Sélection de la langue

Search

Sommaire du brevet 2971729 

Énoncé de désistement de responsabilité concernant l'information provenant de tiers

Une partie des informations de ce site Web a été fournie par des sources externes. Le gouvernement du Canada n'assume aucune responsabilité concernant la précision, l'actualité ou la fiabilité des informations fournies par les sources externes. Les utilisateurs qui désirent employer cette information devraient consulter directement la source des informations. Le contenu fourni par les sources externes n'est pas assujetti aux exigences sur les langues officielles, la protection des renseignements personnels et l'accessibilité.

Disponibilité de l'Abrégé et des Revendications

L'apparition de différences dans le texte et l'image des Revendications et de l'Abrégé dépend du moment auquel le document est publié. Les textes des Revendications et de l'Abrégé sont affichés :

  • lorsque la demande peut être examinée par le public;
  • lorsque le brevet est émis (délivrance).
(12) Brevet: (11) CA 2971729
(54) Titre français: PROMEDICAMENTS DE LA CREATINE, COMPOSITIONS EN CONTENANT ET LEURS PROCEDES D'UTILISATION
(54) Titre anglais: CREATINE PRODRUGS, COMPOSITIONS AND METHODS OF USE THEREOF
Statut: Accordé et délivré
Données bibliographiques
(51) Classification internationale des brevets (CIB):
  • C7D 273/04 (2006.01)
  • A61K 31/4245 (2006.01)
  • A61K 31/5395 (2006.01)
  • A61K 31/551 (2006.01)
  • A61P 25/00 (2006.01)
  • A61P 25/28 (2006.01)
  • C7D 271/07 (2006.01)
  • C7D 273/06 (2006.01)
(72) Inventeurs :
  • BRUBAKER, WILLIAM F. (Etats-Unis d'Amérique)
(73) Titulaires :
  • FARMINGTON PHARMA DEVELOPMENT
(71) Demandeurs :
  • FARMINGTON PHARMA DEVELOPMENT (Etats-Unis d'Amérique)
(74) Agent: DEETH WILLIAMS WALL LLP
(74) Co-agent:
(45) Délivré: 2024-02-20
(86) Date de dépôt PCT: 2015-12-22
(87) Mise à la disponibilité du public: 2016-06-30
Requête d'examen: 2020-10-13
Licence disponible: S.O.
Cédé au domaine public: S.O.
(25) Langue des documents déposés: Anglais

Traité de coopération en matière de brevets (PCT): Oui
(86) Numéro de la demande PCT: PCT/US2015/067283
(87) Numéro de publication internationale PCT: US2015067283
(85) Entrée nationale: 2017-06-20

(30) Données de priorité de la demande:
Numéro de la demande Pays / territoire Date
62/095,295 (Etats-Unis d'Amérique) 2014-12-22

Abrégés

Abrégé français

L'invention concerne des promédicaments de la créatine susceptibles de traverser les membranes, des compositions pharmaceutiques comprenant des promédicaments de la créatine susceptibles de traverser les membranes et des méthodes de traitement de maladies comme l'ischémie, l'insuffisance cardiaque, les troubles neurodégénératifs et les maladies génétiques affectant le système de la créatine kinase, lesdites méthodes demandant l'administration de promédicaments de la créatine ou de compositions pharmaceutiques en contenant. L'invention concerne également le traitement d'une maladie génétique affectant le système de la créatine kinase, comme un trouble du transporteur de la créatine ou un trouble de la synthèse de la créatine, consistant à administrer des promédicaments de la créatine ou des compositions pharmaceutiques en contenant. Des composés de l'invention comprennent des composés de formule (I) ou (III) ou un sel acceptable sur le plan pharmaceutique, un solvate, un tautomère ou un stéréoisomère connexe.


Abrégé anglais


The invention describes membrane permeable creatine prodrugs, pharmaceutical
compositions comprising membrane permeable creatine prodrugs, and methods of
treating
diseases such as ischemia, heart failure, neurodegenerative disorders and
genetic disorders
affecting the creatine kinase system comprising administering creatine
prodrugs or
pharmaceutical compositions thereof. The invention also describes treating a
genetic disease
affecting the creatine kinase system, such as, for example, a creatine
transporter disorder or a
creatine synthesis disorder comprising administering creatine prodrugs or
phannaceutical
compositions thereof. Compounds of the invention include compounds of Formula
(I) or (III),
or a pharmaceutically acceptable salt, solvate, tautomer or stereoisomer
thereof.
<IMG>

Revendications

Note : Les revendications sont présentées dans la langue officielle dans laquelle elles ont été soumises.


CLAIMS
What is claimed is:
1. A compound of Formula (I) or Formula (III), or a pharmaceutically
acceptable
salt, solvate, tautomer or stereoisomer thereof:
wherein the compound of Formula (I) is
HN
R1HN/.\N R1 N N
1bR ir
OH OH
N HN
R1HNN R1 N
[IR
(I)
wherein:
R is ¨CH3 or ¨CD3;
RI is hydrogen, -0R2, -C(0)0R2, ¨C(0)R2,
0 0
0 0
R3 R4
N 0
133
Date recue/Date received 2023-03-10

0 0
R>i 123 R3 R4c0C) csss,
0 0
0
'1\11\10sSS5'
, 0
0 0
R3 R4
0 0 ri
;
n is an integer from 1 to 2;
each R2 is independently hydrogen, Ci_12 alkyl, substituted Ci_12 alkyl, Ci_12
heteroalkyl,
substituted C1-12 heteroalkyl, C3-12 cycloalkyl, substituted C3-12 cycloalkyl,
C4-20 cycloalkylalkyl,
substituted C4_20 cycloalkylalkyl, C4_20 heterocycloalkylalkyl, substituted C4-
20
heterocycloalkylalkyl, C5-12 aryl, substituted C5-12 aryl, C5_12 heteroaryl,
substituted C5-12
heteroaryl, C6_20 arylalkyl, substituted C6_20 arylalkyl, C6_20
heteroarylalkyl or substituted C6-2o
heteroarylalkyl;
each R3 and R4 is independently hydrogen, C1-12 alkyl or substituted C1-12
alkyl;
R23 is hydrogen, Ci_i2 alkyl, substituted Ci_12 alkyl, C5_12 cycloalkyl,
substituted C5-12
cycloalkyl, C5-12 aryl, C5-12 substituted aryl, -C(0)-0R22 or -C(0)-R22;
R22 iS C1-12 alkyl, substituted C1_12 alkyl, C1_12 heteroalkyl, substituted
C1_12 heteroalkyl,
C3-12 cycloalkyl, substituted C3_12 cycloalkyl, C4_20 cycloalkylalkyl,
substituted C4_20
cycloalkylalkyl, C4-20 heterocycloalkylalkyl, substituted C4-20
heterocycloalkylalkyl, C5-12 aryl,
substituted C5_12 aryl, C5_12 heteroaryl, substituted C5_12 heteroaryl, C6_20
arylalkyl, substituted C6_
zo arylalkyl, C6-20 heteroarylalkyl or substituted C6-20 heteroarylalkyl;
-rs48
K iS C1-12 alkyl or substituted CI-12 alkyl; and
wherein each substituent group is independently halogen, -NO2, -OH, -N1-12, -
CN, -CF3, -
OCF3, =0, C1-12 alkyl, C1-12 aikoxy, or -COOR10, wherein R1 ' is hydrogen, C1-
3 alkyl or -
(NR11')2, and wherein each RIF is independently hydrogen or Ci_3 alkyl;
wherein the compound of Foimula (III) is:
134
Date recue/Date received 2023-03-10

0
N H
0
N
(III);
wherein:
W is ¨CH2OH or ¨C(0)0R7;
R is ¨CH3 or ¨CD3;
R.7 is hydrogen, C1-12 alkyl, substituted C1-12 alkyl, C1-12 heteroalkyl,
substituted C1-12
heteroalkyl, C3-12 cycloalkyl, substituted C3-12 cycloalkyl, C4-20
cycloalkylalkyl, substituted C4-20
cycloalkylalkyl, C4_213 heterocycloalkylalkyl, substituted C4_20
heterocycloalkylalkyl, C5-12 aryl,
substituted C5-12 aryl, C5-12 heteroaryl, substituted C5-12 heteroaryl, C6-213
arylalkyl, substituted C6-
20 arylalkyl, C6-20 heteroarylalkyl, substituted C6-20 heteroarylalkyl, -
C(0)R5, -C(0)01e, -
C(0)(NR3R4), -C(R3R4)-C(0)0R22, -C(R3R4)-(0)C(0) R22, -C(R3R4)-(0)C(0)-0R22,
0 0
H N H N
SN /0
N
R3 R4
0 0 or
R3 R4 R23
;
n is an integer from 1 to 2;
each R3 and R4 is independently hydrogen, C1-12 alkyl or substituted C1-12
alkyl; and
135
Date recue/Date received 2023-03-10

R5 is hydrogen, C1-12 alkyl, substituted C1-12 alkyl, C1-12 heteroalkyl,
substituted C1-12
heteroalkyl, C3-12 cycloalkyl, substituted C3-12 cycloalkyl, C4-20
cycloalkylalkyl, substituted C4-20
cycloalkylalkyl, C4_213 heterocycloalkylalkyl, substituted C4_20
heterocycloalkylalkyl, C5-12 aryl,
substituted C5-12 aryl, C5-12 heteroaryl, substituted C5-12 heteroaryl, C6-20
arylalkyl, substituted C6-
20 arylalkyl, C6_20 heteroarylalkyl or substituted C6_20 heteroarylalkyl;
R23 is hydrogen, C1-12 alkyl, substituted C1-12 alkyl, C5-12 cycloalkyl,
substituted C5-12
cycloalkyl, C5_12 aryl, C5-12 substituted aryl, -C(0)-0R22 or -C(0)-R22;
R22 iS C1-12 alkyl, substituted C1-12 alkyl, C1-12 heteroalkyl, substituted C1-
12 heteroa1kyl,
C3-12 cycloalkyl, substituted C3-12 cycloalkyl, C4-2o cycloalkylalkyl,
substituted C4-213
cycloalkylalkyl, C4_213 heterocycloalkylalkyl, substituted C4_20
heterocycloalkylalkyl, C5-12 aryl,
substituted Cs-12 aryl, Cs-12 heteroalyl, substituted C5-12 heteroaryl, C6-20
arylalkyl, substituted C6-
20 arylalkyl, C6-20 heteroarylalkyl or substituted C6-20 heteroarylalkyl; and
wherein each substituent group is independently halogen, -NO2, -OH, -NH2, -CN,
-CF3, -
OCF3, =0, C1-12 alkyl, C1-12 aikoxy, or -COOR', wherein R10' is hydrogen, C1-3
alkyl or -
(NR11')2, and wherein each Riu is independently hydrogen or C1-3 alkyl.
2. The compound of claim 1, wherein each n is independently the integer 1.
3. The compound of claim 1, wherein each n is independently the integer 2.
4. The compound of claim 1, wherein each R2, R5, le, and R22 is
independently C1-6
alkyl, substituted C1-6 alkyl, C3-7 cycloalkyl, substituted C3-7 cycloalkyl,
C5-7 aryl or substituted
C5-7 aryl.
5. The compound of claim 1, wherein each R2, R5, and R7 is independently
hydrogen, methyl, ethyl, n-propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-
butyl, n-pentyl,
isopentyl, sec-pentyl, neopentyl, dodecyl, 1,1-dimethoxyethyl, 1,1 -
diethoxyethyl, phenyl, 4-
methoxyphenyl, benzyl, phenethyl, cyclopropyl, cyclobutyl, cyclopentyl,
cyclohexyl, 2-pyridyl,
3-pyridyl or 4-pyridyl; and
136
Date recue/Date received 2023-03-10

R22 is independently methyl, ethyl, n-propyl, isopropyl, butyl, isobutyl, sec-
butyl, tert-
butyl, n-pentyl, isopentyl, sec-pentyl, neopentyl, dodecyl, 1,1-
dimethoxyethyl, 1,1-diethoxyethyl,
phenyl, 4-methoxyphenyl, benzyl, phenethyl, cyclopropyl, cyclobutyl,
cyclopentyl, cyclohexyl,
2-pyridyl, 3-pyridyl or 4-pyridyl.
6. The compound of claim 1, wherein each R2, R5, and R7 is independently
hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl,
tert-butyl, n-pentyl,
isopentyl, sec-pentyl, neopentyl, dodecyl, 1,1-diethoxyethyl, phenyl,
cyclohexyl or 3-pyridyl;
and R22 is independently methyl, ethyl, n-propyl, isopropyl, n-butyl,
isobutyl, sec-butyl, tert-
butyl, n-pentyl, isopentyl, sec-pentyl, neopentyl, dodecyl, 1,1-diethoxyethyl,
phenyl, cyclohexyl
or 3-pyridyl.
7. The compound of claim 1, wherein each R2, R5, and R7 is independently
hydrogen, methyl, ethyl, n-propyl, isopropyl, dodecyl, tert-butyl, phenyl or
cyclohexyl; and R22
is independently methyl, ethyl, n-propyl, isopropyl, dodecyl, tert-butyl,
phenyl or cyclohexyl.
8. The compound of claim 1, wherein each R2, R5, R7, and R22 is
independently
ethyl, isopropyl or dodecyl.
9. The compound of claim 1, wherein each le and R4 is independently
hydrogen.
10. The compound of claim 1, wherein each R23 is hydrogen, methyl, ethyl, n-
propyl,
isopropyl, tert-butyl, dodecyl, phenyl or cyclohexyl.
11. The compound of claim 1, wherein R23 is methyl.
12. The compound of claim 1, wherein the compound of Formula (I) is a
compound
of Formula (X), Formula (XI), Formula (XII), Formula (XIII), Formula (XIV),
Formula (XV),
Formula (XVa), or Foimula (XVb), or a pharmaceutically acceptable salt,
solvate, tautomer or
stereoisomer thereof;
137
Date recue/Date received 2023-03-10

wherein the compound of Formula (X) is:
HN.....'oN===.....,,o
H 2 N .."*.¨\.N / HN N /
I I
R R
I 1'
0 OH 0 OH
N HN
H 2NN -==I \ __
HN N
I I
R R
(X)
wherein R is ¨CH3 or ¨CD3;
wherein the compound of Foimula (XI) is:
0 0 0 0
N---- "......,;..---"-- HN "...,...,;.------
-------
R240 1 -'==\
N /-...N ,.. ____________________________ 1== __ R240
H
I I
R R
I 1
0 OH
NoOH
HN
R240 1 _ii...._ R240 1
N .\ N / -'==\ ___
N N
H
I I
R R
(XI)
wherein R is ¨CH3 or ¨CD3; and
13 8
Date recue/Date received 2023-03-10

R24 is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl,
phenyl or
cyclohexyl;
wherein the compound of Formula (XII) is:
0
0 0
HN
--===\
R250 N /.."\ N
lb
0 0 OH 0 0
HNOH
--===\ ______________________________________
R250 R250 N
(XII)
wherein R is ¨CH3 or ¨CD3; and
R25 is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl,
phenyl or
cyclohexyl;
wherein the compound of Formula (XIII) is:
139
Date recue/Date received 2023-03-10

0
N /o \/,/o 0
H N
I_ii......
-'==\
R26'N N / R2 6 N N ,...-
H
RI I
R
4 4
0 õ---o-.....,_õ..--=OH 0 H N 0 OH
N
I_ii...... 1
R26N N
R26 N N
H
I I
R R
(XIH)
wherein R is ¨CH3 or ¨CD3; and
R'' is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl,
phenyl or
cyclohexyl;
wherein the compound of Formula (XIV) is:
(:)(:)N
0 0 N
I 1
-\ N N /\. /--N"=.o/\.o/-.''\
N N
I H H
I
R R
(XIV)
wherein R is ¨CH3 or ¨CD3;
wherein the compound of Formula (XV) is:
o o
N o 0 0 N --2:)
I 1
N"'..N/-N.../- \0 ------\N/-\N/
I H H
I
R R
(XV)
wherein R is ¨CH3 or ¨CD3;
140
Date recue/Date received 2023-03-10

wherein the compound of Formula (XVa) is:
o o o o
o 0
R4 R3
N R3 R4 R3g NH R3
I
N /-N 0
..- ¨ N N 0 ..
I H 0 - --= I
0
R R
0 0
,1, 1
0 0
0 0
R3 HOo NH R4
R3
R4 R3
H 0 'c) N R3
N ..'"- N 0
..-----µ -,---
...._ N N 0
I H
I 0
R R
0 --____< 0
0 0
(XVa)
wherein R is ¨CH3 or ¨CD3;
R39 is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl,
phenyl or
cyclohexyl; and
R3 and R4 are each independently hydrogen, C1-12 alkyl or substituted C1-12
alkyl;
wherein the compound of Formula (XVb) is:
o 0
N /- ""--õ. 0 0 0 0
R4 R3 R4 R', HN/
\,
,.._
R--
0 0 /---*-\ X0 ...N/.-t-.N R--/ -,
c,0 0 X0 .....NN/
H I I
R R
I
11
0 0 0 OH 0 0 0 OH
R4 R3, N
..-- R4 R3 HN
R530 0 X0 /.'N /kN)
--%¨
R530 0 X0 ...N..L.Nj
H I I
R R
(XVb)
wherein R is ¨CH3 or ¨CD3;
R3 and le are each independently hydrogen, C1-12 alkyl or substituted C1-12
alkyl; and
R53 is C1_12 alkyl or substituted C1-12 alkyl.
141
Date recue/Date received 2023-03-10

13. The compound of claim 1, wherein the compound of Formula (III) is a
compound
of Formula (XVII), Formula (XVIII), or Formula (XIX), or a pharmaceutically
acceptable salt,
solvate, tautomer or stereoisomer thereof;
wherein the compound of Formula (XVII) is:
0 0
0 0
\\Nr)
R29
0 or
(XVII)
wherein R is ¨CH3 or ¨CD3;
R29 is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl,
phenyl, -
R3 R4
R39
0
cyclohexyl, -CH2-C(0)0R43, -CH2-(0)C(0)R43, -CH2-(0)C(0)0R43 or 0 ;
R39 is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl,
phenyl or
cyclohexyl;
R43 is methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl, phenyl or
cyclohexyl; and
R3 and le are each independently hydrogen, C1-12 alkyl or substituted Ci_12
alkyl;
wherein the compound of Formula (XVIII) is:
0 0
HNNN
--<
o\
0 0
(XVIII)
R is ¨CH3 or ¨CD3;
142
Date recue/Date received 2023-03-10

wherein the compound of Formula (XIX) is:
o o
)----NH HN
O 4
\N\ N/C) \/\ N /0
N
I I
R 0 0 R
(XIX)
wherein R is ¨CH3 or ¨CD3.
14. The compound of
claim 1, which is selected from the group consisting of
0-NH 0-N
0 __ ( 1 0 __ ( * 0 ____ ( .)1
N---- - N '=-C)'''--- N N .-C)- N N ,OH
6D3 0 H 1
0 H 1
CD3 0
/ / /
0-NH
O __ < C)H < 0 0-< 0 ( 1
N N - N N N N (:)"- N----- -
N ''''--=C)'''-
H 1 H H 1 0 C D3 0 0 , 6D3
0
/ / /
0-NH 0-NH
04 0 ____ ( I P-NH
N N (21
______________________ N--_,;-N _,,____OH 0 ( I,,,,
______________________________________________ N N
-''''=
I 0 6D3 1
/ / /
0 ,O,
NH
0-NH 0-NH
0 -( 0 ____ ( 1 N N1H
NN ---''y 0 1\1 - N
CD3 0
D----õD
I 0 D
N
0 ._0,
`--- NH N NH2 "--- N
N '--NH DE) '-N NH2
I D , and I .
,
143
Date recue/Date received 2023-03-10

15. A pharmaceutical composition comprising at least one compound of claims
1 or 14,
and a pharmaceutically acceptable vehicle.
16. The pharmaceutical composition of claim 15, which is in one or more
sustained
release oral dosage forms.
17. The pharmaceutical composition of claim 15, wherein the at least one
compound is
present in:
an amount effective for the treatment of a disease in a patient wherein the
disease is selected
from the group consisting of ischemia, oxidative stress, a neurodegenerative
disease, ischemic
reperfusion injury, a cardiovascular disease, a genetic disease affecting the
creatine kinase system,
multiple sclerosis, a psychotic disorder, and muscle fatigue;
an amount sufficient to effect energy homeostasis in a tissue or an organ
affected by a
disease;
an amount effective for the enhancement of muscle strength in a patient;
an amount effective for the improvement of the viability of a tissue or an
organ; or
an amount effective for the improvement of the viability of cells.
18. The pharmaceutical composition of claim 15, wherein the at least one
compound is
present in an amount effective for the treatment of a genetic disease
affecting the creatine kinase
system.
19. The pharmaceutical composition of claim 15, wherein the at least one
compound is
present in an amount effective for the treatment of a creatine transporter
disorder.
20. The pharmaceutical composition of claim 15, wherein the at least one
compound is
present in an amount effective for the treatment of a creatine synthesis
disorder.
144
Date recue/Date received 2023-03-10

21. Use of a pharmaceutical composition of claim 15 or 16 for treating
ischemia,
oxidative stress, a neurodegenerative disease, ischemic reperfusion injury, a
cardiovascular
disease, a genetic disease affecting the creatine kinase system, multiple
sclerosis, a psychotic
disorder or muscle fatigue.
22. The use of claim 21, wherein the genetic disease affecting the creatine
kinase
system is a creatine transporter disorder or a creatine synthesis disorder.
23. Use of a pharmaceutical composition of claim 15 or 16 for treating a
genetic disease
affecting the creatine kinase system.
24. Use of a pharmaceutical composition of claim 15 or 16 for treating a
creatine
transporter disorder.
25. Use of a pharmaceutical composition of claim 15 or 16 for treating a
creatine
synthesis disorder.
26. Use of a pharmaceutical composition of claim 15 or 16 for enhancing
muscle
strength.
27. Use of a pharmaceutical composition of claim 15 or 16for improving the
viability
of cells.
28. Use of a pharmaceutical composition of claim 15 or 16 for improving the
viability
of a tissue or an organ.
29. Use of a pharmaceutical composition of claim 15 or 16 for effecting
energy
homeostasis in a tissue or an organ.
145
Date recue/Date received 2023-03-10

30. A pharmaceutical composition of claim 15 or 16 for use in treating
ischemia,
oxidative stress, a neurodegenerative disease, ischemic reperfusion injury, a
cardiovascular
disease, a genetic disease affecting the creatine kinase system, multiple
sclerosis, a psychotic
disorder or muscle fatigue.
31. The pharmaceutical composition for use of claim 30, wherein the genetic
disease
affecting the creatine kinase system is a creatine transporter disorder or a
creatine synthesis
disorder.
32. A pharmaceutical composition of claim 15 or 16 for use in treating a
genetic disease
affecting the creatine kinase system.
33. A pharmaceutical composition of claim 15 or 16 for use in treating a
creatine
tansporter disorder.
34. A pharmaceutical composition of claim 15 or 16 for use in treating a
creatine
synthesis disorder.
35. A pharmaceutical composition of claim 15 or 16 for use in enhancing
muscle
strength.
36. A pharmaceutical composition of claim 15 or 16 for use in improving the
viability
of cells.
37. A pharmaceutical composition of claim 15 or 16 for use in improving the
viability
of a tissue or an organ.
38. A pharmaceutical composition of claim 15 or 16 for use in effecting
energy
homeostasis in a tissue or an organ.
146
Date recue/Date received 2023-03-10

Description

Note : Les descriptions sont présentées dans la langue officielle dans laquelle elles ont été soumises.


CREATINE PRODRUGS, COMPOSITIONS AND METHODS OF USE THEREOF
CROSS-REFERENCE TO RELATED APPLICATION
[0001] This application claims the benefit of priority to U.S.
Provisional Application No.
62/095,295, filed on December 22, 2014 and entitled "CREATINE PRODRUGS,
COMPOSITIONS
AND METHODS OF USE THEREOF".
FIELD OF THE INVENTION
[0002] The invention describes membrane permeable creatine prodrugs,
pharmaceutical
compositions comprising membrane permeable creatine prodrugs, and methods of
treating diseases, such
as, for example, ischemia, heart failure, neurodegenerative disorders and
genetic disorders affecting the
creatine kinase system comprising administering creatine prodrugs or
pharmaceutical compositions
thereof. In some embodiments the invention describes treating a genetic
disease affecting the creatine
kinase system, such as, for example, a creatine transporter disorder or a
creatine synthesis disorder
comprising administering creatine prodrugs or pharmaceutical compositions
thereof.
BACKGROUND OF THE INVENTION
[0003] Creatine plays an important part in cellular energy metabolism,
constituting as high-
energy phosphocreatine a significant muscular energy reserve in addition to
adenosine triphosphate
(ATP). In the resting state of the muscle, ATP can transfer a phosphate group
onto creatine, so forming
phosphocreatine, which is then in direct equilibrium with ATP. During muscular
work, it is of vital
importance to replenish ATP stores as rapidly as possible. Phosphocreatine is
available for this purpose
during the first seconds of maximum muscle load; this substance is capable in
a very rapid reaction of
transferring a phosphate group onto adenosine diphosphate by the enzyme
creatine kinase, so reforming
ATP. The creatine kinase system has a dual role in intracellular energy
metabolism-functioning as an
energy buffer to restore depleted ATP levels at sites of high ATP hydrolysis,
and to transferring energy
in the form of phosphocreatine from the mitochondria to other parts of the
cell by a process involving
intermediate energy carriers, several enzymatic reactions, and diffusion
through various intracellular
structures.
[0004] Many pathological disease states arise from a dysfunction in
energy metabolism. Cellular
depletion of ATP stores, as occurs for example during tissue ischemia,
1
Date Recue/Date Received 2022-04-14

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
results in impaired tissue functions and cell death. Of foremost medical
relevance, ischemia-
related cardiovascular disease such as stroke and heart attack remains a
leading cause of
death and morbidity in North America and Europe. Thus, strategies that can
prevent or
reverse ischemia-related tissue damage are expected to have a major impact on
public health.
Energy depletion also contributes to tissue damage during surgery and is a
common cause of
organ transplant failure. Furthermore, reperfusion with oxygen-containing
solutions can
further exacerbate tissue health through production of oxygen radicals.
Therefore, a method
to rapidly restore ATP levels without causing reperfusion injury is likely to
have many
therapeutic applications. Neurodegenerative diseases such as Parkinson's
disease, Alzheimer's
disease, and Huntington's disease are associated with impaired energy
metabolism, and
strategies for improving ATP metabolism could potentially minimize loss of
neurons and
thereby improve the prognosis of patients with these diseases. Finally,
impaired energy
metabolism is an important factor in muscle fatigue and limits physical
endurance Therefore,
a method of preventing or reversing ATP depletion in ischemic or metabolically
active
tissues is likely to have broad clinical utility in a wide range of
indications.
[0005] Creatine supplementation increases intracellular creatine phosphate
levels
(Harris et al., Clinical Sci 1992, 83, 367-74). Creatine readily crosses the
blood-brain barrier
in healthy individuals and brain creatine levels can be increased via oral
administration
(Dechent et al., Am J Physiol 1999, 277, R698-704). Prolonged creatine
supplementation can
elevate the cellular pools of creatine phosphate and increase resistance to
tissue ischemia and
muscle fatigue. Thus, although administration of creatine may have some
therapeutic
usefulness, a modified creatine molecule that is more stable and is more
permeable to barrier
tissues and cellular membranes would have enhanced therapeutic value.
[0006] Creatine prodrugs of the invention are designed to be stable in
biological
fluids, to enter cells by either passive diffusion or active transport, and to
release creatine into
the cellular cytoplasm. Such prodrugs can also cross important barrier tissues
such as the
intestinal mucosa, the blood-brain barrier, and the blood-placental barrier.
Because of the
ability to pass through biological membranes, creatine prodrugs can restore
and maintain
energy homeostasis in ATP depleted cells via the creatine kinase system, and
rapidly restore
ATP levels to protect tissues from further ischemic stress. Creatine prodrugs
having a higher
free energy or lower affinity for creatine kinase, and which can regenerate
ATP under more
severe conditions of energy depletion are also disclosed. Creatine prodrugs of
the invention
can also be used to deliver sustained systemic concentrations of creatine. The
invention is
directed to these, as well as other, important ends.
2

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
SUMMARY OF THE INVENTION
[0007] The present invention relates to membrane permeable creatine
prodrugs,
pharmaceutical compositions comprising the membrane permeable creatine
prodrugs, and
methods of using membrane permeable creatine prodrugs, and pharmaceutical
compositions
thereof. In some embodiments the invention describes treating a genetic
disease affecting the
creatine kinase system, such as, for example, a creatine transporter disorder
or a creatine
synthesis disorder comprising administering creatine prodrugs or
pharmaceutical
compositions thereof.
[0008] In one embodiment, the invention describes compounds of Formula (I) or
a
pharmaceutically acceptable salt, solvate, tautomer or stereoisomer thereof:
wherein the compound of Formula (I) is
0 0 0 0
N HN./
RHNN
RNN
lb
0 0
NOH
HOH
R1HNN R1N
(I)
wherein:
R is ¨CH3 or ¨CD3;
R1 is hydrogen, -0R2, -C(0)0R2, ¨C(0)R2,
0 0
N 0 0
R3 R4
N ><,
0 n 0
3

CA 02971729 2017-06-20
WO 2016/106284 PCT/1JS2015/067283
0 0 R23 R3 R4 0
N 0 0
,0
0 0
R3 R4
R48 X
0 0 n 0
or C5 =
n is an integer from 1 to 2;
each R2 is independently hydrogen, C1-12 alkyl, substituted C1-12 alkyl, C1-12
heteroalkyl, substituted C1-12 heteroalkyl, C3.12 cycloalkyl, substituted
C3.12 cycloalkyl, C4-20
cy cl oalkyl alkyl, substituted C4-20 cyclo al kyl al kyl, C4-20
heterocycloalkylalkyl, substituted C4.
20 heterocycloalkylalkyl, C5-12 aryl, substituted C5-12 aryl, C5-12
heteroaryl, substituted C5-12
heteroaryl, C6.20 arylalkyl, substituted C6-20 arylalkyl, C6-20
heteroarylalkyl or substituted C6-20
heteroarylalkyl;
each R3 and R4 is independently hydrogen, C142 alkyl or substituted Ci_17
alkyl;
R23 is hydrogen, C1-12 alkyl, substituted C1-12 alkyl, C5-12 cycloalkyl,
substituted C5-12
cycloalkyl, C5_12 aryl, and C5-12 substituted aryl, -C(0)-0R22 or -C(0)-R22;
22
K is C1-12 alkyl, substituted C1-12 alkyl, C1-12 heteroalkyl, substituted C1-
12
heteroalkyl, C3-12 cycloalkyl, substituted C3-12 cycloalkyl, C4-20 cy cl oal
kyl alkyl, substituted
C4_20 cycloalkylalkyl, C4_20 heterocycloalkylalkyl, substituted C4_20
heterocycloalkylalkyl, C5_
12 aryl, substituted C5-12 aryl, C5-12 heteroaryl, substituted C5-12
heteroaryl, C6-20 arylalkyl,
substituted C6-20 arylalkyl, C6-20 heteroarylalkyl or substituted C6-20
heteroarylalkyl; and
R48 is C1-12 alkyl or substituted C1-12 alkyl.
[0009] Yet another embodiment describes compounds of Formula (III) or a
pharmaceutically acceptable salt, solvate, tautomer or stereoisomer thereof:
wherein the compound of Formula (III) is:
NH
0

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
wherein:
W is ¨CH2OH or ¨C(0)0R7;
R is ¨CH3 or ¨CD3;
R7 is hydrogen, C1-12 alkyl, substituted C1-12 alkyl, Ci-t2 heteroalkyl,
substituted C1-12
heteroalkyl, C3-12 cycloalkyl, substituted C3-12 cycloalkyl, C4-20
cycloalkylalkyl, substituted
C4-20 cycloalkylalkyl, C4-20 heterocycloalkylalkyl, substituted C4-20
heterocycloalkylalkyl, C5.
12 aryl, substituted C5.12 aryl, C5-12 heteroaryl, substituted C5-12
heteroaryl, C6-20 arylalkyl,
substituted C6_20 arylalkyl, C6.20 heteroarylalkyl, substituted C6_20
heteroarylalkyl, -C(0)R5, -
C(0)0R5, -C(0)(NR3R4), -C(R3R4)-C(0)0R22, -C(R3R4)-(0)C(0) R22, -C(R3R4)-
(0)C(0)-
0R22,
0 0
HN-4
(0 0
`Ni/ LN/
R3 R4
0 R 0 Ror
R3 R4 R23
0
0 ;
n is an integer from 1 to 2,
each le and R4 is independently hydrogen, C1-12 alkyl or substituted C1-12
alkyl, and
R5 is hydrogen, C1-12 alkyl, substituted C1-12 alkyl, C1-12 heteroalkyl,
substituted C1-12
heteroalkyl, C3-12 cycloalkyl, substituted C3.17 cycloalkyl, C4-20
cycloalkylalkyl, substituted
C4.20 cycloalkylalkyl, C4-20 heterocycloalkylalkyl, substituted C4-20
heterocycloalkylalkyl, C5.
12 aryl, substituted C5-12 aryl, C5-12 heteroaryl, substituted C5-12
heteroaryl, C6-20 arylalkyl,
substituted C6.20 arylalkyl, C6.20 heteroarylalkyl or substituted C6_20
heteroarylalkyl;
R23 is hydrogen, C1-12 alkyl, substituted C1-12 alkyl, C5-12 cycloalkyl,
substituted C5-12
cycloalkyl, C512 aryl, and C5-12 substituted aryl, -C(0)-0R22 or -C(0)-R22;
and
R22 is C1-12 alkyl, substituted Ci_12 alkyl, C1-12 heteroalkyl, substituted C1-
12
heteroalkyl, C3-12 cycloalkyl, substituted C3-12 cycloalkyl, C4-20
cycloalkylalkyl, substituted
C4_20 cycl oal kyl alkyl, C4_20 heterocycloal kyl al kyl, substituted C4_20
heterocycloal kyl al kyl, C5_

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
12 aryl, substituted C5-12 aryl, C5-12 heteroaryl, substituted C5-12
heteroaryl, C6-20 arylalkyl,
substituted C6-20 arylalkyl, C6-20 heteroarylalkyl or substituted C6-20
heteroarylalkyl.
[0010] Yet another embodiment describes compounds of Formula (VI) or a
pharmaceutically acceptable salt, solvate, tautomer or stereoisomer thereof:
wherein the compound of Formula (VI) is:
os 12
N
Rio
0
(VI)
wherein:
R is ¨CH3 or ¨CD3;
Rl is hydrogen, C1_12 alkyl, substituted C1.12 alkyl, C1-12 heteroalkyl,
substituted C1-12
heteroalkyl, C3-12 cycloalkyl, substituted C3-12 cycloalkyl, C4-20
cycloalkylalkyl, substituted
C4.20 cycloalkylalkyl, C4-20 heterocycloalkylalkyl, substituted C4-20
heterocycloalkylalkyl, C5.
12 aryl, substituted C5-12 aryl, C5-12 heteroaryl, substituted C5-12
heteroaryl, C6-20 arylalkyl,
substituted C6_20 arylalkyl, C6.20 heteroarylalkyl, substituted C6_20
heteroarylalkyl, -C(0)R5, -
C(0)0R5, -C(0)(NR3R4), -C(R3R4)-C(0)0R22, -C(R3R4)-(0)C(0) R22, -C(R3R4)-
(0)C(0)-
OR22 ;
R11
R3 R4 R23 N
0 R12
0
0 R3 R4 0 R or
HN
R1 2
0 R=
R11 and R'2 are each independently hydrogen or ¨0R13; or and R12
are each -
C(0)R5, with the proviso that both and R12 cannot be hydrogen;
6

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
R13 is independently hydrogen, Ci-12 alkyl, substituted Ci-12 alkyl, C1-12
heteroalkyl,
substituted C1-12 heteroalkyl, C3-12 cycloalkyl, substituted C3-12 cycloalkyl,
C4-20
cycloalkylalkyl, substituted C4-20 cycloalkylalkyl, C4-20
heterocycloalkylalkyl, substituted C4.
20 heterocycloalkylalkyl, C5-12 aryl, substituted C5-12 aryl, C5_12
heteroaryl, substituted C5-12
heteroaryl, C6-20 arylalkyl, substituted C6-20 arylalkyl, C6-20
heteroarylalkyl, substituted C6-20
heteroarylalkyl -CH(0R5), -C(0)R5, -C(0)0R5 or -C(0)(NR3R4);
each R3 and R4 is independently hydrogen, C1-12 alkyl or substituted C1-12
alkyl;
R5 is hydrogen, C1-12 alkyl, substituted C1-12 alkyl, C112 heteroalkyl,
substituted C1.12
heteroalkyl, C3-12 cycloalkyl, substituted C3-12 cycloalkyl, C4-20
cycloalkylalkyl, substituted
C4-20 cycloalkylalkyl, C4-20 heterocycloalkylalkyl, substituted C4.70
heterocycloalkylalkyl, C5.
12 aryl, substituted C5-12 aryl, C5-12 heteroaryl, substituted C5.1/
heteroaryl, C6-20 arylalkyl,
substituted C6_20 arylalkyl, C6_20 heteroarylalkyl or substituted C6_20
heteroarylalkyl;
R23 is hydrogen, C1_12 alkyl, substituted C1-12 alkyl, C542 cycloalkyl,
substituted C5-12
cycloalkyl, C5-12 aryl, and C5-12 substituted aryl, -C(0)-0R22 or -C(0)-R22;
R22
is C1-12 alkyl, substituted C1-12 alkyl, C1-12 heteroalkyl, substituted C1.12
heteroalkyl, C3.12 cycloalkyl, substituted C3.12 cycloalkyl, C4.20
cycloalkylalkyl, substituted
C4-20 cycloalkylalkyl, C4-20 heterocycloalkylalkyl, substituted C4-20
heterocycloalkylalkyl, C5.
12 aryl, substituted C5-12 aryl, C5-12 heteroaryl, substituted C5-12
heteroaryl, C6-20 arylalkyl,
substituted C6-20 arylalkyl, C6-20 heteroarylalkyl or substituted C6-20
heteroarylalkyl; and
n is an integer from 1 to 3.
[0011] One other embodiment describes compounds of Formula (VII) or a
pharmaceutically acceptable salt, solvate, tautomer or stereoisomer thereof:
wherein the compound of Formula (VII) is:
R14
R14
o
NH2
(VII)
wherein:
R is ¨CH3 or ¨CD3;
each R14 is independently hydrogen, C1-12 alkyl, substituted C1.12 alkyl,
C1.12
heteroalkyl, substituted C1-12 heteroalkyl, C3-12 cycloalkyl, substituted C3-
17 cycloalkyl, C4.20
7

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
cycloalkylalkyl, substituted C4-20 cycloalkylalkyl, C4-20
heterocycloalkylalkyl, substituted C4.
20 heterocycloalkylalkyl, C5-12 aryl, substituted C5-12 aryl, C5-12
heteroaryl, substituted C5-12
heteroaryl, C6-20 arylalkyl, substituted C6-20 arylalkyl, C6-20
heteroarylalkyl, substituted C6-20
heteroarylalkyl -CH(0R5), -C(0)R5, -C(0)0R5 or -C(0)(NR3R4),
each R3 and R4 is independently hydrogen, Ci-t2 alkyl or substituted C1-12
alkyl, and
R5 is hydrogen, C1-12 alkyl, substituted C1-12 alkyl, C1-12 heteroalkyl,
substituted C1-12
heteroalkyl, C3-12 cycloalkyl, substituted C1-12 cycloalkyl, C4-20
cycloalkylalkyl, substituted
C4.20 cycloalkylalkyl, C4.20 heterocycloalkylalkyl, substituted C4.20
heterocycloalkylalkyl,
12 aryl, substituted C5-12 aryl, C5-12 heteroaryl, substituted C5-12
heteroaryl, C6-20 arylalkyl,
substituted C6-20 arylalkyl, C6-20 heteroarylalkyl or substituted C6-20
heteroarylalkyl.
[0012] In certain embodiments, the compounds of Formulae (I), (III), (VI),
and (VII),
can include the following features.
[0013] Each R is independently -CH3
[0014] Each R is independently -CD3.
[0015] Each n is independently the integer 1.
[0016] Each n is independently the integer 2.
[0017] Each R2, R5, R6, R7, Rs, R9, R10, R14, R15, -
Kis and R22 is independently C1-6
alkyl, substituted C1-6 alkyl, C3.7 cycloalkyl, substituted C3.7 cycloalkyl,
C5.7 aryl or
substituted C5.7 aryl.
[0018] Each R2, R15, le and R22 is independently
hydrogen, methyl, ethyl, n-propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-
butyl, n-pentyl,
isopentyl, sec-pentyl, neopentyl, dodecyl, 1,1-dimethoxyethyl, 1 , 1-
diethoxyethyl, phenyl, 4-
methoxyphenyl, benzyl, phenethyl, styryl, cyclopropyl, cyclobutyl,
cyclopentyl, cyclohexyl,
2-pyridyl, 3-pyridyl or 4-pyridyl.
[0019] Each R2, R5, R6, R7, Rs, R9, Rio, Ri4, R15, Ris and K-22
is independently
hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl,
tert-butyl, n-pentyl,
isopentyl, sec-pentyl, neopentyl, dodecyl, 1,1 -diethoxyethyl, phenyl,
cyclohexyl or 3-pyridyl
[0020] Each R2, R5, R6, R7, Rs, R9, Rio, Ri4, R15, Ris K
and -22
is independently
hydrogen, methyl, ethyl, n-propyl, isopropyl, dodecyl, tert-butyl, phenyl or
cyclohexyl
[0021] Each R2, Rs, R6, R7, Rs, R9, Rio, Ri4, R15, Ris an K-22
is independently ethyl,
isopropyl or dodecyl
[0022] Each R3 and R4 is independently hydrogen.
[0023] Each R23 is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-
butyl, dodecyl,
phenyl or cyclohexyl.
8

CA 02971729 2017-06-20
WO 2016/106284
PCT/US2015/067283
[0024] Each R23 is methyl.
[0025] Each sub stituent group is independently halogen, -NO2, -OH, -NH2,
-CN, -CF3, -0CF3, =0, C1-12 alkyl, substituted C1-12 alkyl, C1-12 alkoxy or
substituted C1-12
'
alkoxy, -COOR10' wherein R10' is hydrogen, C1.3 alkyl or -(NR11 )2 wherein
each RIF is
independently hydrogen or Ct.3 alkyl.
[0026] In one embodiment, the compound of Formula (I) is a compound of
Formula
(X), Formula (XI), Formula (XII), Formula (XIII), Formula (XIV), Formula (XV),
Formula
(XVa) or Formula (XVb) or a pharmaceutically acceptable salt, solvate,
tautomer or
stereoisomer thereof;
wherein the compound of Formula (X) is:
0 0 0 0
HN
H2N
HN
lb If
NOH
HNOH
H2NN
HN N
(X)
wherein R is ¨CH3 or ¨CD3,
wherein the compound of Formula (XI) is:
9

CA 02971729 2017-06-20
WO 2016/106284 PCT/1JS2015/067283
0 0 0 0
N HN/
'./ N"/"..'
R240 I ______________ li __ R240
H
I I
R R
If 1r
0 0H ,,0 OH
N HN" ..`-'''''
1 I -is, I
R24o R240'\N.7-\.N./ --.1k- --
...,... .......:7".^...N. ...õ...-.
N N
H
I I
R R
(XI)
wherein R is ¨CH3 or ¨CD3, and
R24 is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl,
phenyl or
cyclohexyl;
wherein the compound of Formula (XII) is:
0 N...--,, HN .. 0
.
..---* ---.........:..*0
_....k_
R250-,NN / R25o/'.N\ 1\N
H
I I
R R
1 1
0 0 OH 0 0 OH
N HN
R25o.N/.'N\ N R250 N N
H
I I
R R
(XII)
wherein R is ¨CH3 or ¨CD3, and
R25 is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl,
phenyl or
cyclohexyl;
wherein the compound of Formula (XIII) is.

CA 02971729 2017-06-20
WO 2016/106284
PCT/1JS2015/067283
0 ,, 0 ,..µ4,,?0 0 ,,õ0 .,,,/,
0
N HN
1 ,
_...,,_
R26 N /'.N / R26-N.'',..N /
H
I I
R R
4 1
0 0 OH 0 0 OH
N HN
R.__
=,A".. N./-\ N R26 N
N
H
I I
R R
(XIII)
wherein R is ¨CH3 or ¨CD3; and
R26 is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl,
phenyl or
cyclohexyl;
wherein the compound of Formula (XIV) is:
0 0 0 0
N 0 0 V
N N 0 0 N N
I H H
I
R R
(XIV)
wherein the compound of Formula (XV) is:
N 0 0 N
I N N 0 N N
I H H
I
R R
(XV)
wherein R is ¨CH3 or ¨CD3;
wherein the compound of Formula (XVa) is:
11

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
o R4 o o R39 .., o
R4 R39
o.'oN R3 NH R3
)1.........
N/t\N o
..--=
I 0
R R
0--......<
0 0
0
HOµ\./.'o 0 R39 N
HO\./"'os'NH R4 R39
R3 R4 R3
I 0
R R
0 ---...< 0 -
......<
0 o
(XVa)
wherein R is ¨CH3 or ¨CD3,
R39 is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl,
phenyl or
cyclohexyl; and
R3 and R4 are each independently hydrogen, Ci.12 alkyl or substituted Ci-t2
alkyl;
wherein the compound of Formula (XVb) is:
o N.- ,
R 0 -'o\'o 0 R4 R", 0
4 R3
R
530 ./........oXo/..".....N...1.N
._...._
R53o./'''...\oXoN,,IN.N/'
H
I I
R R
If 41,
0 0o''\./OH 0 0 ..,õ
.............,..OH 0
R4 R3 R4 R3 HN
N I R'i ..-
__ 0 0 ,....,..",,, X 0 N N ..,..."....., ..,..,_
R530/e..0X0/............N. NN/
H
I I
R R
(XVb)
wherein R is ¨CH3 or ¨CD3,
R3 and R4 are each independently hydrogen, C1-12 alkyl or substituted C1-12
alkyl; and
R53 is C1.12 alkyl or substituted C1.12 alkyl.
12

CA 02971729 2017-06-20
WO 2016/106284
PCT/US2015/067283
[0027] In yet another embodiment, the compound of Formula (III) is a
compound of
Formula (XVII), Formula (XVIII) or Formula (XIX) or a pharmaceutically
acceptable salt,
solvate, tautomer or stereoisomer thereof;
wherein the compound of Formula (XVII) is:
0
NH
O\
R29
0
(XVII)
wherein R is ¨CH3 or ¨CD3;
R29 is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl,
phenyl, -
R3 R4
R39
0
0"-=-=.<
cyclohexyl, -CH2-C(0)0R43, -CH2-(0)C(0)R43, -CH2-(0)C(0)0R43 or 0 =
R39 is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl,
phenyl or
cyclohexyl;
R43 is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl,
phenyl or
cyclohexyl; and
R3 and R4 are each independently hydrogen, C1-12 alkyl or substituted CI-L2
alkyl;
wherein the compound of Formula (XVIII) is:
0 0
HN--
./1.k.õ
O\0 0 NN/o
(XVIII)
wherein R is ¨CH3 or ¨CD3;
wherein the compound of Formula (XIX) is:
13

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
0 0
HN--<
\N
0
/N C oh=N
0 0
(XIX)
wherein R is ¨CH5 or ¨CD3.
[0028] In still another embodiment, the compound of Formula (VI) is a
compound of
Formula (XXII), Formula (XXIII), Formula (XXIV), Formula (XXV), Formula
(XXVI),
Formula (XXVII) or Formula (XXVIII) or a pharmaceutically acceptable salt,
solvate,
tautomer or stereoisomer thereof;
wherein the compound of formula (XXII) is:
HO \õ
HN/N
R32
RaR 0
(XXII)
wherein the compound of Formula (XXIII) is:
HO
HN R32
0
(XXIII)
wherein the compound of Formula (XXIV) is:
R330
H2N N R32
0
(XXIV)
wherein the compound of Formula (XXV) is:
14

CA 02971729 2017-06-20
WO 2016/106284
PCT/US2015/067283
NH
HO \.s
HN N R32
0
(XXV)
wherein the compound of Formula (XXVI) is:
R330==.,
HN N R32
0
(XXVI)
wherein the compound of Formula (XXVII) is:
HO
R330
HN N R32
0
(XXVII)
wherein the compound of Formula (XXVIII) is:
R33
N
0
R33JL HN N,//o R32
0
(XXVIII)
wherein R is ¨CH3 or ¨CD3;
le is hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-
butyl;
R32 is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl,
phenyl,
cyclohexyl,

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
R3 R4
R39
0
-CH2-C(0)0R43, -CH2-(0)C(0)R43, -CH2-(0)C(0)0R43 or 0 ;
R39 is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl,
phenyl or
cyclohexyl;
R33 is independently hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl,
dodecyl,
phenyl or cyclohexyl;
R43 is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl,
phenyl or
cyclohexyl; and
R3 and R4 are each independently hydrogen, C1_12 alkyl or substituted C1-12
alkyl.
[0029] In yet another embodiment, the compound of Formula (VII) is a
compound of
Formula (XXIX) or a pharmaceutically acceptable salt, solvate, tautomer or
stereoisomer
thereof;
wherein the compound of Formula (XXIX) is:
R34. R34
0*..."k1H2
(XXIX)
wherein R is -CH3 or -CD3; and
each R34 is independently hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-
butyl,
dodecyl, phenyl or cyclohexyl.
[0030] In one embodiment, the invention describes compounds having the
following
structures:
O-N (D-NH -N 0
N N
N N
H H
0 CD3 0 0
OH
N N N"-y N''
H I H H
CD3 0 0 CD3 0
16

CA 02971729 2017-06-20
WO 2016/106284 PCT/1JS2015/067283
0-NH -NH
0 I 0-NH
--:-. -OH
N N ---
1
I 0 C D3 0 I
0-NH -NH
re -N---õ,,OH
1
CD3 I 8
, ,
--- NH
0-NH N.N.LNH 0.,.0,N
N N NH
1 DD N NH2
C D3 0 I D I
, ,
N_OH N_OH
-.
N I I Boc.,N)LN0
..-.,e.,0õ.,.=
N N ,
DDNH2 Boc, H I 6 H I
D CD3 0
HO, HO, '.N1
0 N 0 N
0 0,N
OAN,K,N,-.=y0
0,1,N-1õNi-0
0AN.)N,,,=OH
H I H 1
0 ,,..---...... C D3 0 ) H I 0
0 03,/
00,N 3,/
0........ N 0 IN
0 NN,.,=,y0H
N N H2
N N H2 i
) H CD3 0 I or CD3 .
, ,
or a pharmaceutically acceptable salt, solvate, tautomer or stereoisomer
thereof.
[0031] In
another embodiment, the invention describes pharmaceutical compositions
comprising a therapeutically effective amount of at least one compound of
Formulae (I), (III),
(VI), and (VII), and any subgenera or species thereof, or a pharmaceutically
acceptable salt,
solvate, tautomer or stereoisomer thereof, or a pharmaceutically acceptable
solvate of any of
the foregoing, and a pharmaceutically acceptable vehicle. In one embodiment,
the invention
describes pharmaceutical compositions comprising a therapeutically effective
amount of at
least one compound as disclosed herein, or a pharmaceutically acceptable salt,
solvate,
tautomer or stereoisomer thereof, or a pharmaceutically acceptable solvate of
any of the
foregoing, and a pharmaceutically acceptable vehicle
17

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
[0032] In some embodiments, the pharmaceutical compositions can be
formulated in
one or more sustained release oral dosage forms.
[0033] In one embodiment, the pharmaceutical composition comprises at least
one
compound of the present invention in an amount effective for the treatment of
a disease in a
patient wherein the disease is ischemia, oxidative stress, a neurodegenerative
disease,
ischemic reperfusion injury, a cardiovascular disease, a genetic disease
affecting the creatine
kinase system, multiple sclerosis, a psychotic disorder, and muscle fatigue;
an amount
sufficient to effect energy homeostasis in a tissue or an organ affected by a
disease; an
amount effective for the enhancement of muscle strength in a patient; an
amount effective for
the improvement of the viability of a tissue or an organ, or an amount
effective for the
improvement of the viability of cells In another embodiment, the
pharmaceutical
composition comprises at least one compound of the present invention in an
amount effective
for the treatment of a genetic disease affecting the creatine kinase system.
In some
embodiments, the pharmaceutical composition comprises at least one compound of
the
present invention in an amount effective for the treatment of a creatine
transporter disorder.
In one embodiment, the pharmaceutical composition comprises at least one
compound of the
present invention in an amount effective for the treatment of a creatine
synthesis disorder.
[0034] In one embodiment the invention describes methods for treating a
disease in a
patient associated with a dysfunction in energy metabolism such as ischemia,
oxidative stress,
a neurodegenerative disease, including amyotrophic lateral sclerosis (ALS),
Huntington's
disease, Parkinson's disease or Alzheimer's disease, ischemic reperfusion
injury, a
cardiovascular disease, multiple sclerosis (MS), a psychotic disorder, a
genetic disease
affecting the creatine kinase system or muscle fatigue in a patient comprising
administering
to a patient in need of such treatment a therapeutically effective amount of
at least one
compound of Formulae (I), (III), (VI), (VII), and any subgenera or species
thereof, or a
pharmaceutically acceptable salt, solvate, tautomer or stereoisomer thereof,
or a
pharmaceutical composition comprising at least one compound of Formulae (I),
(III), (VI),
(VII), and any subgenera or species thereof, or a pharmaceutically acceptable
salt, solvate,
tautomer or stereoisomer thereof.
[0035] In another embodiment, methods are described for treating a genetic
disease
affecting the creatine kinase system, such as, for example, a creatine
transporter disorder or a
creatine synthesis disorder in a patient comprising administering to a patient
in need of such
treatment a therapeutically effective amount of at least one compound of
Formulae (I), (III),
(VI), (VII), and any subgenera or species thereof, or a pharmaceutically
acceptable salt,
18

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
solvate, tautomer or stereoisomer thereof, or a pharmaceutical composition
comprising at
least one compound of Foimulae (I), (III), (VI), (VII), and any subgenera or
species thereof,
or a pharmaceutically acceptable salt, solvate, tautomer or stereoisomer
thereof.
[0036] In further embodiments, methods are described for enhancing muscle
strength
in a patient comprising administering to a patient in need of such enhancement
a
therapeutically effective amount of at least one compound of Formulae (I),
(III), (VI), (VII),
and any subgenera or species thereof, or a pharmaceutically acceptable salt,
solvate, tautomer
or stereoisomer thereof, or a pharmaceutical composition comprising at least
one compound
of Formulae (I), (III), (VI), (VII), and any subgenera or species thereof, or
a pharmaceutically
acceptable salt, solvate, tautomer or stereoisomer thereof.
[0037] In yet one more embodiment, methods are described for increasing the
viability of a tissue or an organ comprising contacting the tissue or the
organ with an
effective amount of at least one compound of Formulae (I), (III), (VI), (VII),
and any
subgenera or species thereof, or a pharmaceutically acceptable salt, solvate,
tautomer or
stereoisomer thereof, or a pharmaceutical composition comprising at least one
compound of
Formulae (I), (III), (VI), (VII), and any subgenera or species thereof, or a
phaimaceutically
acceptable salt, solvate, tautomer or stereoisomer thereof.
[0038] In still one more embodiment, methods are described for improving
the
viability of isolated cells comprising contacting the cells with an effective
amount of at least
one compound of Formulae (I), (III), (VI), (VII), and any subgenera or species
thereof, or a
pharmaceutically acceptable salt, solvate, tautomer or stereoisomer thereof,
or a
pharmaceutical composition comprising at least one compound of Formulae (I),
(III), (VI),
(VII), and any subgenera or species thereof, or a pharmaceutically acceptable
salt, solvate,
tautomer or stereoisomer thereof.
[0039] In another embodiment, methods are described for treating a disease
associated with oxidative stress are provided comprising administering to a
patient in need of
such treatment an effective amount of at least one compound of Formulae (I),
(III), (VI),
(VII), and any subgenera or species thereof, or a pharmaceutically acceptable
salt, solvate,
tautomer or stereoisomer thereof, or a pharmaceutical composition comprising
at least one
compound of Formulae (I), (III), (VI), (VII), and any subgenera or species
thereof, or a
pharmaceutically acceptable salt, solvate, tautomer or stereoisomer thereof.
[0040] In one more embodiment, methods for improving the viability of a
tissue or an
organ are described for treating a tissue or organ manifesting a dysfunction
in energy
metabolism are provided comprising contacting at least one compound of
Formulae (I), (III),
19

(VI), (VII), and any subgenera or species thereof, or a pharmaceutically
acceptable salt, solvate, tautomer
or stereoisomer thereof, or a pharmaceutical composition comprising at least
one compound of Formulae
(I), (III), (VI), (VII), and any subgenera or species thereof, or a
pharmaceutically acceptable salt, solvate,
tautomer or stereoisomer thereof, with the tissue or organ.
[0041] In yet one more embodiment, methods are described for effecting
energy homeostasis in
a tissue or an organ are provided comprising contacting at least one compound
of Formulae (I), (III),
(VI), (VII), and any subgenera or species thereof, or a pharmaceutically
acceptable salt, solvate, tautomer
or stereoisomer thereof, or a pharmaceutical composition comprising at least
one compound of Formulae
(I), (III), (VI), (VII), and any subgenera or species thereof, or a
pharmaceutically acceptable salt, solvate,
tautomer or stereoisomer thereof, with the tissue or the organ.
[0042] In another embodiment, methods are described for treating an
oxidatively stressed tissue
or organ are provided comprising contacting at least one compound of Formulae
(I), (III), (VI), (VII),
and any subgenera or species thereof, or a pharmaceutically acceptable salt,
solvate, tautomer or
stereoisomer thereof, or a pharmaceutical composition comprising at least one
compound of Formulae
(I), (III), (VI), (VII), and any subgenera or species thereof, or a
pharmaceutically acceptable salt, solvate,
tautomer or stereoisomer thereof, with the tissue or organ.
[0043] Unless otherwise defined, all technical and scientific terms used
herein have the same
meaning as commonly understood by one of ordinary skill in the art to which
this invention belongs. In
the specification, the singular forms also include the plural unless the
context clearly dictates otherwise.
Although methods and materials similar or equivalent to those described herein
can be used in the
practice or testing of the present invention, suitable methods and materials
are described below. In the
case of conflict, the present specification, including definitions, will
control. In addition, the materials,
methods and examples are illustrative only and are not intended to be
limiting.
[0044] Other features and advantages of the invention will be apparent
from the following
detailed description and claims.
Date Recue/Date Received 2022-04-14

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
DETAILED DESCRIPTION
Definitions
[0045] A dash ("-") that is not between two letters or symbols is used to
indicate a
point of attachment for a moiety or substituent. For example, -CONH2 is
attached through the
carbon atom.
[0046] "Alkyl" by itself or as part of another substituent refers to a
saturated or
unsaturated, branched or straight-chain, monovalent hydrocarbon radical
derived by the
removal of one hydrogen atom from a single carbon atom of a parent alkane,
alkene or
alkyne. Examples of alkyl groups include, but are not limited to, methyl;
ethyls such as
ethanyl, ethenyl, and ethynyl; propyls such as propan-l-yl, propan-2-yl, prop-
1-en-1-yl, prop-
1-en-2-yl, prop-2-en-1-y1 (allyl), prop-1-yn-1-yl, prop-2-yn-1-yl, etc.;
butyls such as butan-l-
yl, butan-2-y1 , 2-m ethyl-propan-l-yl, 2-methyl -propan-2-yl, but-l-en-l-yl ,
but-l-en-2-yl, 2-
methyl-prop-1-en-1-yl, but-2-en- 1 -yl, but-2-en-2-yl, buta-1,3 -di en- 1 -yl,
buta-1,3-dien-2-yl,
but-l-yn-l-yl, but-1-yn-3-yl, but-3-yn-1-yl, etc.; and the like.
[0047] The term "alkyl" is specifically intended to include groups having
any degree
or level of saturation, i.e., groups having exclusively single carbon-carbon
bonds, groups
having one or more double carbon-carbon bonds, groups having one or more
triple carbon-
carbon bonds, and groups having mixtures of single, double, and triple carbon-
carbon bonds.
Where a specific level of saturation is intended, the terms "alkanyl,"
"alkenyl," and "alkynyl"
are used. In certain embodiments, an alkyl group can have from 1 to 20 carbon
atoms, in
certain embodiments, from 1 to 12 carbon atoms, in certain embodiments, from 1
to 10
carbon atoms, in certain embodiments, from 1 to 6 carbon atoms, and in certain
embodiments, from 1 to 3 carbon atoms.
[0048] "Alkoxy" by itself or as part of another substituent refers to a
radical OR31
where Rn- is chosen from alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl,
cycloalkylalkyl,
heterocycloalkylalkyl, aryl, heteroaryl, arylalkyl, and heteroarylalkyl, as
defined herein
Examples of alkoxy groups include, but are not limited to, methoxy, ethoxy,
propoxy,
butoxy, cyclohexyloxy, and the like.
[0049] "Aryl" by itself or as part of another substituent refers to a
monovalent
aromatic hydrocarbon radical derived by the removal of one hydrogen atom from
a single
carbon atom of a parent aromatic ring system. Aryl encompasses 5- and 6-
membered
carbocyclic aromatic rings, for example, benzene; bicyclic ring systems
wherein at least one
ring is carbocyclic and aromatic, for example, naphthalene, indane, and
tetralin; and tricyclic
ring systems wherein at least one ring is carbocyclic and aromatic, for
example, fluorene.
21

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
Aryl encompasses multiple ring systems having at least one carbocyclic
aromatic ring fused
to at least one carbocyclic aromatic ring, cycloalkyl ring or heterocycloalkyl
ring. For
example, aryl includes 5- and 6-membered carbocyclic aromatic rings fused to a
5- to 7-
membered heterocycloalkyl ring containing one or more heteroatoms chosen from
N, 0, and
S. For such fused, bicyclic ring systems wherein only one of the rings is a
carbocyclic
aromatic ring, the point of attachment may be at the carbocyclic aromatic ring
or the
heterocycloalkyl ring. Examples of aryl groups include, but are not limited
to, groups derived
from aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene,
benzene,
chrysene, coronene, fluoranthene, fluorene, hexacene, hexaphene, hexylene, as-
indacene, s-
indacene, indane, indene, naphthalene, octacene, octaphene, octalene, ovalene,
penta-2,4-
diene, pentacene, pentalene, pentaphene, perylene, phenalene, phenanthrene,
picene,
pleiadene, pyrene, pyranthrene, rubicene, triphenylene, trinaphthalene, and
the like. In certain
embodiments, an aryl group can have from 6 to 20 carbon atoms, from 6 to 12
carbon atoms,
and in certain embodiments, from 6 to 8 carbon atoms. Aryl, however, does not
encompass or
overlap in any way with heteroaryl, separately defined herein.
[0050] "Arylalkyl" by itself or as part of another substituent refers to
an acyclic alkyl
radical in which one of the hydrogen atoms bonded to a carbon atom, typically
a terminal or
sp3 carbon atom, is replaced with an aryl group. Examples of arylalkyl groups
include, but are
not limited to, benzyl, 2-phenylethan-1-yl, 2-phenylethen-1-yl,
naphthylmethyl, 2-
naphthyl ethan-l-yl, 2-naphthylethen-1-yl, naphthobenzyl, 2-naphthophenylethan-
1-y1 and the
like. Where specific alkyl moieties are intended, the nomenclature
arylalkanyl, arylalkenyl or
arylalkynyl is used. In certain embodiments, an arylalkyl group is Co
arylalkyl, e.g., the
alkanyl, alkenyl or alkynyl moiety of the arylalkyl group is C<sub>1-10</sub> and
the aryl moiety is
C6-20, in certain embodiments, an arylalkyl group is C6.20arylalkyl, e.g., the
alkanyl, alkenyl or
alkynyl moiety of the arylalkyl group is C1.8 and the aryl moiety is C6.1/.
[0051] "AUC" is the area under a curve representing the concentration of a
compound or metabolite thereof in a biological fluid in a patient as a
function of time
following administration of the compound to the patient In certain
embodiments, the
compound can be a prodrug and the metabolite can be a drug. Examples of
biological fluids
include plasma and blood. The AUC may be determined by measuring the
concentration of a
compound or metabolite thereof in a biological fluid such as the plasma or
blood using
methods such as liquid chromatography-tandem mass spectrometry (LC/MS/MS), at
various
time intervals, and calculating the area under the plasma concentration-versus-
time curve.
Suitable methods for calculating the AUC from a drug concentration-versus-time
curve are
22

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
well known in the art. As relevant to the invention, an AUC for a drug or
metabolite thereof
may be determined by measuring over time the concentration of the drug in the
plasma, blood
or other biological fluid or tissue of a patient following administration of a
corresponding
compound of the invention to the patient.
[0052] "Bioavailability" refers to the rate and amount of a drug that
reaches the
systemic circulation of a patient following administration of the drug or
prodrug thereof to
the patient and can be determined by evaluating, for example, the plasma or
blood
concentration-versus-time profile for a drug. Parameters useful in
characterizing a plasma or
blood concentration-versus-time curve include the area under the curve (AUC),
the time to
maximum concentration (Tmax), and the maximum drug concentration (Cmax), where
C. is
the maximum concentration of a drug in the plasma or blood of a patient
following
administration of a dose of the drug or form of drug to the patient, and Tim,
is the time to the
maximum concentration (Cmax) of a drug in the plasma or blood of a patient
following
administration of a dose of the drug or form of drug to the patient.
[0053] "Cmax" is the maximum concentration of a drug in the plasma or
blood of a
patient following administration of a dose of the drug or prodrug to the
patient.
[0054] "Tmax" is the time to the maximum (peak) concentration (Cmax) of a
drug in the
plasma or blood of a patient following administration of a dose of the drug or
prodrug to the
patient.
[0055] "Compounds of the invention" or "compound of the invention",
include any
specific compounds within these formulae. Compounds may be identified either
by their
chemical structure and/or chemical name. When the chemical structure and
chemical name
are conflicting, the chemical structure is determinative of the identity of
the compound. The
compounds described herein may comprise one or more chiral centers and/or
double bonds
and therefore may exist as stereoisomers such as double-bond isomers (i.e.,
geometric
isomers), enantiomers or diastereomers. Accordingly, any chemical structures
within the
scope of the specification depicted, in whole or in part, with a relative
configuration
encompass all possible enantiomers and stereoisomers of the illustrated
compounds including
the stereoisomerically pure form (e.g., geometrically pure, enantiomerically
pure or
diastereomerically pure) and enantiomeric and stereoisomeric mixtures.
Enantiomeric and
stereoisomeric mixtures may be resolved into their component enantiomers or
stereoisomers
using separation techniques or chiral synthesis techniques well known to the
skilled artisan.
Compounds of the invention are also referred to as "prodrugs of creatine" or
"prodrugs of the
invention."
23

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
[0056] Compounds of invention include, but are not limited to,
stereoisomers or
optical isomers of compounds of the invention, racemates thereof, and other
mixtures thereof.
In such embodiments, the single enantiomers or diastereomers, i.e., optically
active forms,
can be obtained by asymmetric synthesis or by resolution of the racemates.
Resolution of the
racemates may be accomplished, for example, by conventional methods such as
crystallization in the presence of a resolving agent or chromatography, using,
for example a
chiral high-pressure liquid chromatography (HPLC) column. In addition,
compounds of the
invention include Z- and E-forms (or cis- and trans-forms) of compounds with
double bonds.
In embodiments in which compounds of the invention exist in various tautomeric
forms, the
compounds include all tautomeric forms of the compound.
[0057] A "stereoisomer" refers to a compound made up of the same atoms
bonded by
the same bonds but having different three-dimensional structures, which are
not
interchangeable. The present invention contemplates various stereoisomers and
mixtures
thereof and includes "enantiomers", which refers to two stereoisomers whose
molecules are
nonsuperimposable mirror images of one another.
[0058] Compounds of the invention may also exist in several tautomeric
forms, and
the depiction herein of one tautomer is for convenience only, and is also
understood to
encompass other tautomers of the form shown. Accordingly, the chemical
structures depicted
herein encompass all possible tautomeric forms of the illustrated compounds.
The term
"tautomer" as used herein refers to isomers that change into one another with
great ease so
that they can exist together in equilibrium. For example, ketone and enol are
two tautomeric
forms of one compound. In another example, a substituted 1,2,4-triazole
derivative may exist
in at least three tautomeric forms as shown below:
R-ri RT2 RT2
RT2 N NrN,
RT1 is H or optionally substituted alkyl, r¨N N_
sN N,// N RT2 is an optionally substituted aryl.
R-ri
[0059] Compounds of the invention also include isotopically labeled
compounds
where one or more atoms have an atomic mass different from the atomic mass
conventionally
found in nature. Examples of isotopes that may be incorporated into the
compounds disclosed
herein include, but are not limited to, 2H, 3H, itc, 13C, 14C, 15N, 180, 170
etc. Compounds
may exist in unsolvated forms as well as solvated forms, including hydrated
forms and as N-
oxides. In general, compounds may be hydrated, solvated or N-oxides. Certain
compounds
may exist in multiple crystalline or amorphous forms. Compounds of the
invention include
24

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
pharmaceutically acceptable salts thereof or phaimaceutically acceptable
solvates of the free
acid form of any of the foregoing, as well as crystalline forms of any of the
foregoing.
[0060] "Creatine kinase system" includes, but is not limited to the
creatine
transporter, creatine, creatine kinase, creatine phosphate, and the
intracellular energy
transport of creatine, creatine kinase, and/or creatine phosphate. The
creatine kinase system
includes mitochondrial and cytoplasmic creatine kinase systems. Affecting the
creatine
kinase system refers to the transport, synthesis, metabolism, translocation,
and the like, of the
compounds and proteins comprising the creatine kinase system.
[0061] "Cycloalkyl" by itself or as part of another substituent refers to
a saturated or
partially unsaturated cyclic alkyl radical. Where a specific level of
saturation is intended, the
nomenclature "cycloalkanyl" or "cycloalkenyl" is used. Examples of cycloalkyl
groups
include, but are not limited to, groups derived from cyclopropane,
cyclobutane, cyclopentane,
cyclohexane, and the like. In certain embodiments, a cycloalkyl group is C3-15
cycloalkyl, C5_
12 cycloalkyl, and in certain embodiments, C3_7 cycloalkyl.
[0062] "Cycloalkylalkyl" by itself or as part of another substituent
refers to an
acyclic alkyl radical in which one of the hydrogen atoms bonded to a carbon
atom, typically a
terminal or sp3 carbon atom, is replaced with a cycloalkyl group. Where
specific alkyl
moieties are intended, the nomenclature cycloalkylalkanyl, cycloalkylalkenyl
or
cycloalkylalkynyl is used. In certain embodiments, a cycloalkylalkyl group is
C7-30
cycloalkylalkyl, e.g., the alkanyl, alkenyl or alkynyl moiety of the
cycloalkylalkyl group is
C1_10 and the cycloalkyl moiety is C6.20, and in certain embodiments, a
cycloalkylalkyl group
is C7.20 cycloalkylalkyl, e.g., the alkanyl, alkenyl or alkynyl moiety of the
cycloalkylalkyl
group is CI-8 and the cycloalkyl moiety is C4-20 Or C6-12.
[0063] "Disease" refers to a disease, disorder, condition, symptom or
indication.
[0064] "Halogen" refers to a fluoro, chloro, bromo or iodo group.
[0065] "Heteroalkyl" by itself or as part of another substituent refer to
an alkyl group
in which one or more of the carbon atoms (and any associated hydrogen atoms)
are each
independently replaced with the same or different heteroatomic groups.
Examples of
heteroatomic groups include, but are not limited to, -0-, -S-, -0-0-, -S-S-, -
0-S-, -NR57R58-,
=N-N=, -N=N-, -N=N-NR59R60, _pR61_, _P(0)2-, -P0R62-, -0-P(0)2-, -SO-, -SO2-, -
SnR63R64-
, and the like, where R57, R58, R59, R60, R61, R62,
R63, and R64 are each independently chosen
from hydrogen, C1-12 alkyl, substituted C1-12 alkyl, C6-12 aryl, substituted
C6-12 aryl, C7-18
arylalkyl, substituted C748 arylalkyl, C3.7 cycloalkyl, substituted C3.7
cycloalkyl, C3.7
heterocycloalkyl, substituted C3.7 heterocycloalkyl, C1-12 heteroalkyl,
substituted C1.12

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
heteroalkyl, C6.12 heteroaryl, substituted C6.'2 heteroaryl, C2.18
heteroarylalkyl or substituted
C7.18 heteroarylalkyl. Where a specific level of saturation is intended, the
nomenclature
"heteroalkanyl," "heteroalkenyl," or R60, R61, R62, R61,
and R64 "heteroalkynyl" is used. In
certain embodiments, R57, R58, R59, are each independently chosen from
hydrogen and C1.3
alkyl.
[0066] "Heteroaryl" by itself or as part of another substituent refers to
a monovalent
heteroaromatic radical derived by the removal of one hydrogen atom from a
single atom of a
parent heteroaromatic ring system. Heteroaryl encompasses multiple ring
systems having at
least one heteroaromatic ring fused to at least one other ring, which can be
aromatic or non-
aromatic. Heteroaryl encompasses 5- to 7-membered aromatic, monocyclic rings
containing
one or more, for example, from 1 to 4 or in certain embodiments, from 1 to 3,
heteroatoms
chosen from N, 0, and S, with the remaining ring atoms being carbon; and
bicyclic
heterocycloalkyl rings containing one or more, for example, from 1 to 4 or in
certain
embodiments, from 1 to 3, heteroatoms chosen from N, 0, and S, with the
remaining ring
atoms being carbon and wherein at least one heteroatom is present in an
aromatic ring. For
example, heteroaryl includes a 5- to 7-membered heteroaromatic ring fused to a
5- to 7-
membered cycloalkyl ring. For such fused, bicyclic heteroaryl ring systems
wherein only one
of the rings contains one or more heteroatoms, the point of attachment may be
at the
heteroaromatic ring or the cycloalkyl ring. In certain embodiments, when the
total number of
N, S. and 0 atoms in the heteroaryl group exceeds one, the heteroatoms are not
adjacent to
one another. In certain embodiments, the total number of N, 5, and 0 atoms in
the heteroaryl
group is not more than two. In certain embodiments, the total number of N, S,
and 0 atoms in
the aromatic heterocycle is not more than one. Heteroaryl does not encompass
or overlap
with aryl as defined herein.
[0067] Examples of heteroaryl groups include, but are not limited to,
groups derived
from acri dine, arsindol e, carbazol e, 13-carb ol in e, chrom an e, chrom en
e, cinnoline, furan,
imidazole, indazole, indole, indoline, indolizine, isobenzofuran, isochromene,
isoindole,
isoindoline, isoquinoline, isothiazole, isoxazole, naphthyridine, oxadiazole,
oxazole,
perimidine, phenanthridine, phenanthroline, phenazine, phthalazine, pteridine,
purine, pyran,
pyrazine, pyrazole, pyridazine, pyridine, pyrimidine, pyrrole, pyrrolizine,
quinazoline,
quinoline, quinolizine, quinoxaline, tetrazole, thiadiazole, thiazole,
thiophene, triazole,
xanthene, and the like. In certain embodiments, a heteroaryl group is from 5-
to 20-membered
heteroaryl, in certain embodiments from 5- to 10-membered heteroaryl, and in
certain
embodiments from 6- to 8-heteroaryl. In certain embodiments heteroaryl groups
are those
26

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
derived from thiophene, pyrrole, benzothiophene, benzofuran, indole, pyridine,
quinoline,
imidazole, oxazole or pyrazine.
[0068] "Heteroarylalkyl" by itself or as part of another sub stituent
refers to an acyclic
alkyl radical in which one of the hydrogen atoms bonded to a carbon atom, is
replaced with a
heteroaryl group. Typically a terminal or sp3 carbon atom is the atom replaced
with the
heteroaryl group. Where specific alkyl moieties are intended, the nomenclature
"heteroarylalkanyl," "heteroarylalkenyl," and "heterorylalkynyl" is used. In
certain
embodiments, a heteroarylalkyl group is a 6- to 30-membered heteroarylalkyl,
e.g., the
alkanyl, alkenyl or alkynyl moiety of the heteroarylalkyl is 1- to 10-membered
and the
heteroaryl moiety is a 5- to 20-membered heteroaryl, and in certain
embodiments, 6- to 20-
membered heteroarylalkyl, e.g., the alkanyl, alkenyl or alkynyl moiety of the
heteroarylalkyl
is 1- to 8-membered and the heteroaryl moiety is a 5- to 12-membered
heteroaryl.
[0069] "Heterocycloalkyl" by itself or as part of another substituent
refers to a
partially saturated or unsaturated cyclic alkyl radical in which one or more
carbon atoms (and
any associated hydrogen atoms) are each independently replaced with the same
or different
heteroatom. Examples of heteroatoms to replace the carbon atom(s) include, but
are not
limited to, N, P, 0, S, Si, etc. Where a specific level of saturation is
intended, the
nomenclature "heterocycloalkanyl" or "heterocycloalkenyl" is used. Examples of
heterocycloalkyl groups include, but are not limited to, groups derived from
epoxides,
azirines, thiiranes, imidazolidine, morpholine, piperazine, piperidine,
pyrazolidine,
pyrrolidine, quinuclidine, and the like.
[0070] "Heterocycloalkylalkyl" by itself or as part of another sub
stituent refers to an
acyclic alkyl radical in which one of the hydrogen atoms bonded to a carbon
atom, typically a
terminal or sp3carbon atom, is replaced with a heterocycloalkyl group. Where
specific alkyl
moieties are intended, the nomenclature heterocycloalkylalkanyl,
heterocycloalkylalkenyl or
heterocycloalkylalkynyl is used. In certain embodiments, a
heterocycloalkylalkyl group is a
6- to 30-membered heterocycloalkylalkyl, e.g., the alkanyl, alkenyl or alkynyl
moiety of the
heterocycloalkylalkyl is 1- to 10-membered and the heterocycloalkyl moiety is
a 5- to 20-
membered heterocycloalkyl, and in certain embodiments, 6- to 20-membered
heterocycloalkylalkyl, e.g., the alkanyl, alkenyl or alkynyl moiety of the
heterocycloalkylalkyl is 1- to 8-membered and the heterocycloalkyl moiety is a
5- to 12-
m emb ered heterocycloalkyl.
[0071] "Leaving group" refers to an atom or a group capable of being
displaced by a
nucleophile and includes halogen, such as chloro, bromo, fluoro, and iodo,
alkoxycarbonyl
27

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
(e.g., acetoxy), aryloxycarbonyl, me syl oxy, tosyloxy,
trifluoromethanesulfonyloxy, aryloxy
(e.g., 2,4-dinitrophenoxy), methoxy, N,0-dimethylhydroxylamino, and the like.
[0072] "Parent aromatic ring system" refers to an unsaturated cyclic or
polycyclic ring
system having a conjugated t electron system. Included within the definition
of "parent
aromatic ring system" are fused ring systems in which one or more of the rings
are aromatic
and one or more of the rings are saturated or unsaturated, such as, for
example, fluorene,
indane, indene, phenalene, etc. Examples of parent aromatic ring systems
include, but are not
limited to, aceanthrylene, acenaphthylene, acephenanthrylene, anthracene,
azulene, benzene,
chrysene, coronene, fluoranthene, fluorene, hexacene, hexaphene, hexylene, as-
indacene, s-
indacene, indane, indene, naphthalene, octacene, octaphene, octalene, ovalene,
penta-2,4-
diene, pentacene, pentalene, pentaphene, perylene, phenalene, phenanthrene,
picene,
pleiadene, pyrene, pyranthrene, rubicene, triphenylene, trinaphthalene, and
the like.
[0073] "Parent heteroaromatic ring system" refers to an aromatic ring
system in which
one or more carbon atoms (and any associated hydrogen atoms) are each
independently
replaced with the same or different heteroatom. Examples of heteroatoms to
replace the
carbon atoms include, but are not limited to, N, P, 0, S, and Si, etc.
Specifically included
within the definition of "parent heteroaromatic ring systems" are fused ring
systems in which
one or more of the rings are aromatic and one or more of the rings are
saturated or
unsaturated, such as, for example, arsindole, benzodioxan, benzofuran,
chromane, chromene,
indole, indoline, xanthene, etc. Examples of parent heteroaromatic ring
systems include, but
are not limited to, arsindole, carbazole, 13-carboline, chromane, chromene,
cinnoline, furan,
imidazole, indazole, indole, indoline, indolizine, isobenzofuran, isochromene,
isoindole,
isoindoline, isoquinoline, isothiazole, isoxazole, naphthyridine, oxadiazole,
oxazole,
perimidine, phenanthridine, phenanthroline, phenazine, phthalazine, pteridine,
purine, pyran,
pyrazine, pyrazole, pyridazine, pyridine, pyrimidine, pyrrole, pyrrolizine,
quinazoline,
quinoli ne, quinol izine, quinoxaline, tetrazol e, thi adiazol e, thiazol e,
thi ophene, tri azol e,
xanthene, and the like.
[0074] "Patient" refers to animals, preferably mammals, most preferably
humans, and
includes males and females, and children and adults.
[0075] "Phaimaceutical composition" refers to at least one compound of the
invention
and at least one pharmaceutically acceptable vehicle, with which the at least
one compound
of the invention is administered to a patient, contacted with a tissue or
organ or contacted
with a cell. "Pharmaceutically acceptable" refers to approved or approvable by
a regulatory
28

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
agency of the Federal or a state government or listed in the U.S.
Phatmacopoeia or other
generally recognized pharmacopoeia for use in animals, and more particularly
in humans.
[0076]
"Pharmaceutically acceptable salt" refers to a salt of a compound, which
possesses the desired pharmacological activity of the parent compound. Such
salts include:
(1) acid addition salts, formed with inorganic acids such as hydrochloric
acid, hydrobromic
acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed
with organic acids
such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic
acid, glycolic acid,
pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic
acid, fumaric acid,
tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl)benzoic acid,
cinnamic acid,
mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethane-
disulfonic acid, 2-
hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic
acid, 2-
naphthal en e sul foni c acid, 4-toluenesulfonic acid,
camphorsul fonic acid, 4-
methylbicyclo[2.2.2]-oct-2-ene-1-carboxylic acid, glucoheptonic acid, 3-
phenylpropionic
acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid,
gluconic acid,
glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic
acid, and the like,
and (2) salts formed when an acidic proton present in the parent compound is
replaced by a
metal ion, e.g., an alkali metal ion, an alkaline earth ion or an aluminum
ion; or coordinates
with an organic base such as ethanolamine, diethanolamine, triethanolamine, N-
methylglucamine, and the like. In certain embodiments, a pharmaceutically
acceptable salt is
the hydrochloride salt.
[0077]
"Pharmaceutically acceptable vehicle" refers to a pharmaceutically acceptable
diluent, a pharmaceutically acceptable adjuvant, a pharmaceutically acceptable
excipient, a
pharmaceutically acceptable carrier or a combination of any of the foregoing
with which a
compound of the invention may be administered to a patient and which does not
destroy the
pharmacological activity thereof and which is non-toxic when administered in
doses
sufficient to provide a therapeutically effective amount of the compound.
[0078]
"Prodrug" refers to a derivative of a drug molecule that requires a
transformation within the body to release the active drug Prodrugs are
frequently, although
not necessarily, pharmacologically inactive until converted to the parent
drug. Compounds of
Formulae (I), (III), (VI), (VII), and any subgenera or species thereof, are
prodrugs of creatine
that can be metabolized within a patient's body to release creatine.
[0079]
"Promoiety" refers to a group bonded to a drug, typically to a functional
group
of the drug, via bond(s) that are cleavable under specified conditions of use.
The bond(s)
between the drug and promoiety may be cleaved by enzymatic or non-enzymatic
means.
29

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
Under the conditions of use, for example following administration to a
patient, the bond(s)
between the drug and promoiety may be cleaved to release the parent drug. The
cleavage of
the promoiety may proceed spontaneously, such as via a hydrolysis reaction or
it may be
catalyzed or induced by another agent, such as by an enzyme, by light, by acid
or by a change
of or exposure to a physical or environmental parameter, such as a change of
temperature,
pH, etc. The agent may be endogenous to the conditions of use, such as an
enzyme present in
the systemic circulation of a patient to which the prodrug is administered or
the acidic
conditions of the stomach or the agent may be supplied exogenously.
[0080] "Protecting group" refers to a grouping of atoms, which when
attached to a
reactive group in a molecule masks, reduces or prevents that reactivity.
Examples of
protecting groups can be found in Wuts and Greene, "Protective Groups in
Organic
Synthesis," John Wiley & Sons, 4th ed. 2006; Harrison et al., "Compendium of
Organic
Synthetic Methods," Vols. 1-11, John Wiley & Sons 1971-2003; Larock
"Comprehensive
Organic Transformations," John Wiley & Sons, 2nd ed. 2000; and Paquette,
"Encyclopedia of
Reagents for Organic Synthesis," John Wiley & Sons, 11th ed. 2003. Examples of
amino
protecting groups include, but are not limited to, formyl, acetyl,
trifluoroacetyl, benzyl,
benzyloxycarbonyl (CBZ), tert-butoxycarbonyl (Boc), trimethylsilyl (TMS), 2-
trimethylsilyl-
ethanesulfonyl (SES), trityl and substituted trityl groups, allyloxycarbonyl,
9-
fluorenylmethyloxycarbonyl (FMOC), nitro-veratryloxycarbonyl (NVOC), and the
like.
Examples of hydroxy protecting groups include, but are not limited to, those
in which the
hydroxy group is either acylated or alkylated such as benzyl, and trityl
ethers as well as alkyl
ethers, tetrahydropyranyl ethers, trialkylsilyl ethers, and allyl ethers.
[0081] "Solvate" refers to a molecular complex of a compound with one or
more
solvent molecules in a stoichiometric or non-stoichiometric amount. Such
solvent molecules
are those commonly used in the pharmaceutical art, which are known to be
innocuous to
recipient, e.g., water, ethanol, and the like. A molecular complex of a
compound or moiety of
a compound and a solvent can be stabilized by non-covalent intra-molecular
forces such as,
for example, electrostatic forces, van der Waals forces or hydrogen bonds. The
term
"hydrate" refers to a complex in which the one or more solvent molecules are
water including
monohydrates and hemi-hydrates.
[0082] "Substantially one diastereomer" refers to a compound containing
two or more
stereogenic centers such that the diastereomeric excess (d.e.) of the compound
is greater than
or about at least 90%. In certain embodiments, the d.e. is, for example,
greater than or at least

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
about 91%, about 92%, about 93%, about 940/0, about 95%, about 96%, about 97%,
about
98% or about 99%.
[0083] "Substituted" refers to a group in which one or more hydrogen atoms
are each
independently replaced with the same or different substituent(s). Examples of
substituents
include, but are not limited to, -M, -R70, -0-, =0, -OW , -SR.7 , -5-, =S, -
NR70R71, =NR70, -
CF3, -CN, -OCN, -SCN, -NO, -NO2, =N2, -N3, -S(0)20-, -S(0)20H, -S(0)2R70, -
0S(02)0-, -
OS(0)2R7 , -P(0)(0-)2, -P(0)(0R70)(0), -0P(0)(0R70)(0R71), -C(0)R70, -C(S)R70,
-
C(0)0R70, -C(0)NR7 R71, -C(0)0-, -C(S)0R70, -NR72C(0)NR70R71, -NR72C(S)NR7
R11, -
mcc (N-R13)N-R70-K71,
and -C(NR72)NR70R 71where M is independently a halogen; R7 , R71,
R72, and R73 are each independently chosen from hydrogen, alkyl, alkoxy,
cycloalkyl,
heterocycloalkyl, aryl, and heteroaryl or R70and WI-together with the nitrogen
atom to which
they are bonded form a ring chosen from a heterocycloalkyl ring In certain
embodiments,
R70, R71, R72, and R73are each independently chosen from hydrogen, C1_6 alkyl,
C1_6 alkoxy,
C3_12 cycloalkyl, C3_12 heterocycloalkyl, C6_12 aryl, and C6_17 heteroaryl. In
certain
embodiments, each substituent is independently selected from halogen, -OH, -
CN, -CF3, =0,
-NO2, Ci.3 alkoxy, Ci.3 alkyl, -COOR8 wherein R8 is slected from hydrogen,
C1.3 alkyl and
(NR74)2 wherein each R74 is independently hydrogen or Ci.3 alkyl.
[0084] In certain embodiments, substituted aryl and substituted heteroaryl
include one
or more of the following substitute groups: F, Cl, Br, Ci.3 alkyl, substituted
alkyl, Ci.3 alkoxy,
-S(0)2NR50R5I, -NR50R5I, -CF3, -0CF3, -CN, -NR505(0)2R51, -NR50C(0)R51, C5.10
aryl,
substituted C5.10 aryl, C5-10 heteroaryl, substituted C5.10 heteroaryl, -
C(0)0R50, -NO2, -
C(0)R50, -C(0)NR50R51, -OCHF2, C1.3 acyl, -5R50, -5(0) 20H, -S(0)2R50, -
S(0)R50, -
C(S)R50, -C(0)0-, -C(S)0R50, -NR50C(0)NR51R52, -NR50C(S)NR51R52, and -
C(NR50)NR51R52, C3.8 cycloalkyl, and substituted C3.8 cycloalkyl, wherein R50,
R51, and R52
are each independently selected from hydrogen and C1.4 alkyl.
[0085] In certain embodiments, a substituent group can be selected from
halogen, -
NO2, -OH, -COOH, -1\1112, -CN, -CF3, -0CF3, C1_8 alkyl, substituted Cis alkyl,
C1_8 alkoxy,
and substituted C1_8 alkoxy, wherein the each substituent of the substituted
C1_8 alkyl and C1-8
alkoxy is independently selected from halogen, -NO2, -OH, -COOH, -NH2, -CN, -
CF3, -
OCF3.
[0086] In certain embodiments, each substituent is independently selected
from
halogen, -OH, -CN, -CF3, =0, -NO2, C1.3 alkoxy, C1.3 alkyl, -COOW wherein le
is slected
from hydrogen, C1.3 alkyl and (NR74)2 wherein each R74 is independently
hydrogen or C1.3
alkyl.
31

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
[0087] "Therapeutically effective amount" refers to the amount of a
compound that,
when administered to a subject for treating a disease or disorder or at least
one of the clinical
symptoms of a disease or disorder, is sufficient to affect such treatment of
the disease,
disorder or symptom. The "therapeutically effective amount" can vary
depending, for
example, on the compound, the disease, disorder, and/or symptoms of the
disease or disorder,
severity of the disease, disorder, and/or symptoms of the disease or disorder,
the age, weight,
and/or health of the patient to be treated, and the judgment of the
prescribing physician. An
appropriate amount in any given instance can be readily ascertained by those
skilled in the art
or capable of determination by routine experimentation.
[0088] "Therapeutically effective dose" refers to a dose that provides
effective
treatment of a disease or disorder in a patient. A therapeutically effective
dose may vary from
compound to compound, and from patient to patient, and may depend upon factors
such as
the condition of the patient and the route of delivery. A therapeutically
effective dose may be
determined in accordance with routine pharmacological procedures known to
those skilled in
the art.
[0089] "Treating" or "treatment" of any disease or disorder refers to
arresting or
ameliorating a disease, disorder or at least one of the clinical symptoms of a
disease or
disorder, reducing the risk of acquiring a disease, disorder or at least one
of the clinical
symptoms of a disease or disorder, reducing the development of a disease,
disorder or at least
one of the clinical symptoms of the disease or disorder or reducing the risk
of developing a
disease or disorder or at least one of the clinical symptoms of a disease or
disorder.
"Treating" or "treatment" also refers to inhibiting the disease or disorder,
either physically,
(e.g., stabilization of a discernible symptom), physiologically, (e.g.,
stabilization of a physical
parameter) or both, and to inhibiting at least one physical parameter that may
or may not be
discernible to the patient. In certain embodiments, "treating" or "treatment"
refers to delaying
the onset of the disease or disorder or at least one or more symptoms thereof
in a patient
which may be exposed to or predisposed to a disease or disorder even though
that patient
does not yet experience or display symptoms of the disease or disorder.
[0090] Reference is now made in detail to certain embodiments of
compounds,
compositions, and methods. The disclosed embodiments are not intended to be
limiting of the
claims. To the contrary, the claims are intended to cover all alternatives,
modifications, and
equivalents.
32

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
Creatine Prodrugs
[0091] In certain embodiments, a creatine prodrug is a compound of Formula
(I) or a
pharmaceutically acceptable salt, solvate, tautomer or stereoisomer thereof:
wherein the compound of Formula (I) is
0 (:)
HN''
I
R1HN.-'---'\N/
RiN N
I I
R R
l l
0 OH 0 OH
N''/. '=-'-' HN''' '-'''/.
--.1k¨ ...õ.1"........ ....õ---
R1HN N RiN N
I I
R R
(I)
wherein:
R is ¨CH3 or ¨CD3;
R' is hydrogen, -0R2, -C(0)0R2, ¨C(0)R2,
o'-:,=.,,,/o \,
N 0 A 0
1 R3 R-
0
IH
R,
m 3
R23 IA R4 0
N 0 0
1 's n Ociss
NNOc_S'55' C)i_-0
IH
0 , 0
R3 R-
Ra8 .........,....0 0 X 0 ...../..,.........,õ5.....
n
or c53 .
,
33

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
n is an integer from 1 to 2,
each R2 is independently hydrogen, C1.12 alkyl, substituted C1-12 alkyl, C1-12
heteroalkyl, substituted C1-12 heteroalkyl, C3-12 cycloalkyl, substituted C3-
12 cycloalkyl, C4.20
cy cl oalkyl alkyl, substituted C4.20 cyclo al kyl al kyl, C4.20
heterocycloalkylalkyl, substituted C4.
20 heterocycloalkylalkyl, C5-12 aryl, substituted C5-12 aryl, C5-12
heteroaryl, substituted C5-12
heteroaryl, C6-20 arylalkyl, substituted C6-20 arylalkyl, C6-20
heteroarylalkyl or substituted C6-20
heteroarylalkyl;
each R3 and R4 is independently hydrogen, C142 alkyl or substituted Ci.12
alkyl;
R23 is hydrogen, C1-12 alkyl, substituted C1-12 alkyl, C5-12 cycloalkyl,
substituted C5-12
cycloalkyl, C5-12 aryl, and C5-12 substituted aryl, -C(0)-0R22 or -C(0)-R22;
R22 is C1-12 alkyl, substituted Ci_u alkyl, C1-12 heteroalkyl, substituted C1-
12
heteroalkyl, C3_12 cycloalkyl, substituted C3_12 cycl oalkyl, C4_20 cycl oal
kyl al kyl, substituted
C4_20 cycloalkylalkyl, C4_20 heterocycloalkylalkyl, substituted C4_20
heterocycloalkylalkyl, C5_
12 aryl, substituted C5-12 aryl, C5-12 heteroaryl, substituted C5-12
heteroaryl, C6-20 arylalkyl,
substituted C6.20 arylalkyl, C6.20 heteroarylalkyl or substituted C6.20
heteroarylalkyl; and
R48 is C1-12 alkyl or substituted C1.12 alkyl.
[0092] In certain embodiments of a compound of Formula (I), n is the
integer 1.
[0093] In certain embodiments of a compound of Formula (I), n is the
integer 2.
[0094] In certain embodiments of a compound of Formula (I) each R2 and R22
is
independently hydrogen, methyl, ethyl, n-propyl, isopropyl, butyl, isobutyl,
sec-butyl, tert-
butyl, n-pentyl, isopentyl, sec-pentyl, neopentyl, dodecyl, 1, 1 -
dimethoxyethyl, 1,1 -
diethoxyethyl, phenyl, 4-methoxyphenyl, benzyl, phenethyl, styryl,
cyclopropyl, cyclobutyl,
cyclopentyl, cyclohexyl, 2-pyridyl, 3 -pyridyl or 4-pyridyl.
[0095] In certain embodiments of a compound of Formula (I), each R2 and R22
is
independently hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl,
sec-butyl, tert-
butyl, n-pentyl, isopentyl, sec-pentyl, neopentyl, dodecyl, 1,1 -
diethoxyethyl, phenyl,
cyclohexyl or 3 -pyri dyl .
[0096] In certain embodiments of a compound of Formula (I), each R2 and R22
is
independently independently hydrogen, methyl, ethyl, n-propyl, isopropyl,
dodecyl, tert-
butyl, phenyl or cyclohexyl.
[0097] In certain embodiments of a compound of Formula (I), each R2 and R22
is
independently ethyl, isopropyl or dodecyl.
[0098] In certain embodiments of a compound of Formula (I), R3 and R4 each
independently is hydrogen.
34

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
[0099] In certain embodiments of a compound of Formula (I), R23 is
hydrogen,
methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl, phenyl or cyclohexyl.
[00100] In certain embodiments of a compound of Formula (I), R2' is methyl.
[00101] In certain embodiments of a compound of Formula (I), each
substituent group
is independently halogen, -NO2, -OH, -NH2, -CN, -CF3, -0CF3, =0, C1-12 alkyl,
substituted
C1-12 alkyl, C1-12 alkoxy or substituted C1-12 alkoxy, -COOR1 ' wherein RI is
hydrogen, C1-3
alkyl or -(NR11')2 wherein each Rir is independently hydrogen or C1.2 alkyl.
[00102] In some embodiment, the compound of Formula (I) is a compound of
Formula
(X), Formula (XI), Formula (XII), Formula (XIII), Formula (XIV), Formula (XV),
Formula
(XVa) or Formula (XVb) or a pharmaceutically acceptable salt, solvate,
tautomer or
stereoisomer thereof;
wherein the compound of Formula (X) is:
0 0 0 0
H N
H2N N HN,/\
lb
NA../'OH
HNOH
H2N N HNe"..\
(X)
wherein R is ¨CH3 or ¨CD3,
wherein the compound of Formula (XI) is:

CA 02971729 2017-06-20
WO 2016/106284 PCT/1JS2015/067283
0 0 0 0
HN,/ \, ,,,,
N''' N''.",
R240 D24n
-....mk- 1 x ...,
N\ N N / N\ N.,:5=.N.//
H
I I
R R
1 1
0 OH 0 OH
N HN
R24o'.\N R240 .7.-N "=-=...., ....,"%.......
......./0"
N N
H
I I
R R
(XI)
wherein R is ¨CH3 or ¨CD3, and
R24 is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl,
phenyl or
cyclohexyl;
wherein the compound of Formula (XII) is:
0 N...--,, HN ... 0
.
_....,k_
R250.,N N / R25o/'.N\ 1\N
H
I I
R R
l 1
0 0 OH 0 0 OH
N HN
R25o. N N R250 N N
H
I I
R R
(XII)
wherein R is ¨CH3 or ¨CD3, and
R25 is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl,
phenyl or
cyclohexyl;
wherein the compound of Formula (XIII) is.
36

CA 02971729 2017-06-20
WO 2016/106284 PCT/1JS2015/067283
0
N /o`=,."-o
H N
1 ,
R26. N N / R26'''....s.. N. N /
H
I I
R R
l If
0
No'.7'OH 0 0
R__
=,AN R.....N
9A''''N\ N-"'"\. N
H
1 I
R R
(XIII)
wherein R is ¨CH3 or ¨CD3; and
- 26
K is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl, phenyl
or
cyclohexyl;
wherein the compound of Formula (XIV) is:
oo N 0 0 N
N N 0 0 N N
I H H
I
R R
(XIV)
wherein R is ¨CH3 or ¨CD3;
wherein the compound of Formula (XV) is:
N 0 0 N
o'oN'
I N N 0 N N
I H H
I
R R
(XV)
wherein R is ¨CH3 or ¨CD3;
wherein the compound of Formula (XVa) is:
37

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
o.'oN 0
R3 R4 R39 0,.._ _,0%._
-NH 0 R3 R4 R39
N N 0 ..---
R R
0----_< 0--......<
0 0
HO./'oN 0 R39
R3 R3 HOµ\./"'os-NH 0
R4 R39 R4
1 I I 0
R R
0--...< 0--.....
0 o
(XVa)
wherein R is ¨CH3 or ¨CD3;
R39 is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl,
phenyl or
cyclohexyl, and
R3 and R4 is independently hydrogen, C1-12 alkyl or substituted C1-12 alkyl;
wherein the compound of Formula (XVb) is:
o , R", HN ., F , o IR
_,,,,o
r IR' N '
R5300Xc) N jN,- _1...._
--.4-
R53ooXo.N1-1N/
H I I
R R
11
0 , 0 H
N.. 0 0 H
HN,*'/e
R" R3 R4 R3
R53ooXoN,JLN)
-.."'- Fz53o..0X0..N. N./
H I I
R R
(XVb)
wherein R is ¨CH3 or ¨CD3;
R3 and R4 are each independently hydrogen, C112 alkyl or substituted C1-12
alkyl; and
R53 is C1-12 alkyl or substituted C1_17 alkyl.
[00103] In certain embodiments in the compounds of Formula (XI), (XII) and
(XIII),
each R24, R25 and R26 is independently ethyl, isopropyl or dodecyl.
[00104] In certain embodiments in the compounds of Formula (XVa), R39 is
methyl,
ethyl, n-propyl, isopropyl, tert-butyl, dodecyl, phenyl or cyclohexyl.
38

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
[00105] In certain embodiments in the compounds of Formula (XVa), R39 is
methyl,
[00106] In certain embodiments in the compounds of Formula (XVa) or (XVb),
R3 and
R4 are each hydrogen.
[00107] In certain embodiments in the compounds of Formula (XVb), R'3 is
methyl,
ethyl, n-propyl, isopropyl or tert-butyl.
[00108] In certain embodiments a creatine prodrug is a compound of Formula
(III) or
a pharmaceutically acceptable salt, solvate, tautomer or stereoisomer thereof:
wherein the compound of Formula (III) is:
NH
N N
vµi
wherein:
W is ¨CH2OH or ¨C(0)0R7;
R is ¨CH3 or ¨CD3;
R7 is hydrogen, C1-12 alkyl, substituted C1-12 alkyl, C1-12 heteroalkyl,
substituted C1-12
heteroalkyl, C3-12 cycloalkyl, substituted C3-12 cycloalkyl, C4-20
cycloalkylalkyl, substituted
C4.20 cycloalkylalkyl, C4-20 heterocycloalkylalkyl, substituted C4-20
heterocycloalkylalkyl, C.
12 aryl, substituted C5-12 aryl, C5-12 heteroaryl, substituted C5-12
heteroaryl, C6-20 arylalkyl,
substituted C6.20 arylalkyl, C6.20 heteroarylalkyl, substituted C6.20
heteroarylalkyl, -C(0)R5, -
C(0)0R5, -C(0)(NR3R4), -C(R3R4)-C(0)0R22, -C(R3R4)-(0)C(0) R22, -C(R3R4)-
(0)C(0)-
OR22,
0 0
HN
HN-4
0
c.S5N N
R3 R4
0 0 or
39

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
R3 R4 R23
0
0 ;
n is an integer from 1 to 2;
each R3 and R4 is independently hydrogen, C 1- t2 alkyl or substituted C1-12
alkyl; and
R5 is hydrogen, C1.17 alkyl, substituted C1.12 alkyl, C1-12 heteroalkyl,
substituted C1.12
heteroalkyl, C3.12 cycloalkyl, substituted C3-12 cycloalkyl, C4-20
cycloalkylalkyl, substituted
C4-20 cycloalkylalkyl, C4.20 heterocycloalkylalkyl, substituted C4.20
heterocycloalkylalkyl, C.
12 aryl, substituted C5.12 aryl, C5_12 heteroaryl, substituted C5-12
heteroaryl, C6.20 arylalkyl,
substituted C6.20 arylalkyl, C6-20 heteroarylalkyl or substituted C6-20
heteroarylalkyl;
R23 is hydrogen, C1.12 alkyl, substituted C1-12 alkyl, C5-12 cycloalkyl,
substituted C5-12
cycloalkyl, C5-12 aryl, and C5-12 substituted aryl, -C(0)-0R22 or -C(0)-R22;
and
R22 is C1-17 alkyl, substituted C1-12 alkyl, C1-12 heteroalkyl, substituted C1-
12
heteroalkyl, C3-12 cycloalkyl, substituted C3-12 cycloalkyl, C4-20
cycloalkylalkyl, substituted
C4.20 cycloalkylalkyl, C4.20 heterocycloalkylalkyl, substituted C4.20
heterocycloalkylalkyl, C5.
12 aryl, substituted C5-12 aryl, C5-12 heteroaryl, substituted C5-12
heteroaryl, C6.20 arylalkyl,
substituted C6.20 arylalkyl, C6-20 heteroarylalkyl or substituted C6.70
heteroarylalkyl.
[00109] In certain embodiments of a compound of Formula (III), n is the
integer 1.
[00110] In certain embodiments of a compound of Formula (III), n is the
integer 2.
[00111] In certain embodiments of a compound of Formula (III), each R5, R7
and R22 is
independently C1.6 alkyl, substituted C1.6 alkyl, C3-7 cycloalkyl, substituted
C3-7 cycloalkyl,
C5-7 aryl or substituted C5.7 aryl.
[00112] In certain embodiments of a compound of Formula (III), each R5, R7
and R22 is
independently hydrogen, methyl, ethyl, n-propyl, isopropyl, butyl, isobutyl,
sec-butyl, tert-
butyl, n-pentyl, isopentyl, sec-pentyl, neopentyl, dodecyl, 1, 1-
dimethoxyethyl, 1,1-
diethoxyethyl, phenyl, 4-methoxyphenyl, benzyl, phenethyl, styryl,
cyclopropyl, cyclobutyl,
cycl op entyl , cycl oh exyl , 2-pyridyl, 3 -pyri dyl or 4-pyri dyl .
[00113] In certain embodiments of a compound of Formula (III), each R5, R7
and R22 is
independently hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl,
sec-butyl, tert-
butyl, n-pentyl, isopentyl, sec-pentyl, neopentyl, dodecyl, 1,1 -
diethoxyethyl, phenyl,
cyclohexyl or 3 -pyridyl.

CA 02971729 2017-06-20
WO 2016/106284
PCT/US2015/067283
[00114] In certain embodiments of a compound of Formula (III), each R5, R7
and R22 is
independently hydrogen, methyl, ethyl, n-propyl, isopropyl, dodecyl, tert-
butyl, phenyl or
cyclohexyl.
[00115] In certain embodiments of a compound of Formula (III), each R5, R7
and R22 is
independently ethyl, isopropyl or dodecyl.
[00116] In certain embodiments of a compound of Formula (III), each R3 and
R4 is
independently hydrogen.
[00117] In certain embodiments of a compound of Formula (III), each R23 is
hydrogen,
methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl, phenyl or cyclohexyl.
[00118] In certain embodiments of a compound of Formula (III), each R23 is
methyl.
[00119] In certain embodiments of a compound of Formula (III), each each
substituent
group is independently halogen, -NO2, -OH, -NH2, -CN, -CF3, -0CF3, =0, C1_12
alkyl,
substituted Ci_12 alkyl, C1_12 alkoxy or substituted C1_12 alkoxy, -COOR1 '
wherein RI . is
hydrogen, C1_3 alkyl or E.11)2 N
wherein each R" is independently hydrogen or C1_3 alkyl.
¨
[00120] In yet another embodiment, the compound of Formula (III) is a
compound of
Formula (XVII), Formula (XVIII) or Formula (XIX) or a pharmaceutically
acceptable salt,
solvate, tautomer or stereoisomer thereof;
wherein the compound of Formula (XVII) is:
NH
0
0
\N)
R29
0
(XVII)
wherein R is ¨CH3 or
R29 is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl,
phenyl, -
R3 R4
R39
cyclohexyl, -CH2-C(0)0R43, -CH2-(0)C(0)R43, -CH2-(0)C(0)0R43 or 0 ;
R39 is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl,
phenyl or
cyclohexyl;
41

CA 02971729 2017-06-20
WO 2016/106284 PCT/1JS2015/067283
R43 is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl,
phenyl or
cyclohexyl; and
R3 and R4 are each independently hydrogen, Ci-12 alkyl or substituted C1-12
alkyl;
wherein the compound of Formula (XVIII) is:
0 0
Y.-NH HN
0 0
\
NN
0 0
(XVIII)
wherein R is ¨CH3 or ¨CD3;
wherein the compound of Formula (XIX) is:
0 0
Y.-NH
0 0
\ 0 =., /
NN
0 0
(XIX)
wherein R is ¨CH3 or ¨CD3.
[00121] In certain embodiments of a compound of Formula (XVII), each R29
and R43 is
independently ethyl, isopropyl or dodecyl.
[00122] In certain embodiments of a compound of Formula (XVII), each R39 is
methyl.
In certain embodiments of a compound of Formula (XVII) R' and R4 are each
hydrogen.
[00123] In certain embodiments a creatine prodrug is a compound of Formula
(VI) or a
pharmaceutically acceptable salt, solvate, tautomer or stereoisomer thereof:
wherein the compound of Formula (VI) is:
Di
0
(VI)
wherein:
R is ¨CH3 or ¨CD3;
42

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
R1 is hydrogen, C1-12 alkyl, substituted C1,2 alkyl, C1-12 heteroalkyl,
substituted C1.12
heteroalkyl, C3-12 cycloalkyl, substituted C3-12 cycloalkyl, C4-20
cycloalkylalkyl, substituted
C4.20 cycloalkylalkyl, C4-20 heterocycloalkylalkyl, substituted C4-20
heterocycloalkylalkyl, C5.
12 aryl, substituted C5-12 aryl, C5-12 heteroaryl, substituted C5.12
heteroaryl, C6-20 arylalkyl,
substituted C6-20 arylalkyl, C6-20 heteroarylalkyl, substituted C6.20
heteroarylalkyl, -C(0)R5, -
C(0)0R5, -C(0)(NR3R4), -C(R3R4)-C(0)0R22, -C(R4R4)-(0)C(0) R22, -C(R3R4)-
(0)C(0)-
OR22 ;
R11
R3 R4 R23
R12
0 R3 R4 0 R or
Ri
HN
0
RH and R12 are each independently hydrogen or ¨0R13; or RH and R12 are each -
C(0)R5, with the proviso that both R11 and R12 cannot be hydrogen
R13 is independently hydrogen, C1-12 alkyl, substituted Ci-12 alkyl, C1.12
heteroalkyl,
substituted C1-12 heteroalkyl, C3-12 cycloalkyl, substituted C3-12 cycloalkyl,
C4-20
cycloalkylalkyl, substituted C4-20 cycloalkylalkyl, C4-20
heterocycloalkylalkyl, substituted C4.
20 heterocycloalkylalkyl, C5-12 aryl, substituted C5-12 aryl, C5-12
heteroaryl, substituted C5-12
heteroaryl, C6-20 arylalkyl, substituted C6.20 arylalkyl, C6-20
heteroarylalkyl, substituted C6.20
heteroarylalkyl -CH(OR5), -C(0)R5, -C(0)0R5 or -C(0)(NR3R4);
each le and R4 is independently hydrogen, Ci_t, alkyl or substituted C1.32
alkyl;
R5 is hydrogen, C1-12 alkyl, substituted Ci.17 alkyl, C1-12 heteroalkyl,
substituted C1-17
heteroalkyl, C3-12 cycloalkyl, substituted C3_17 cycloalkyl, C4-20
cycloalkylalkyl, substituted
C4.20 cycl alkyl alkyl, C4.20 heterocycl alkyl alkyl, substituted C4.20
heterocycl alkyl alkyl, C5.
12 aryl, substituted C5_12 aryl, C5_12 heteroaryl, substituted C5_12
heteroaryl, C6_20 aryl alkyl ,
substituted C6-20 arylalkyl, C6-20 heteroarylalkyl or substituted C6-20
heteroarylalkyl;
R23 is hydrogen, C1-12 alkyl, substituted C1-12 alkyl, C5-12 cycloalkyl,
substituted C5-12
cycloalkyl, C5-12 aryl, and C5-12 substituted aryl, -C(0)-0R22 or -C(0)-R22,
43

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
R22 is C1-12 alkyl, substituted C1-12 alkyl, C1-12 heteroalkyl, substituted C1-
12
heteroalkyl, C3-12 cycloalkyl, substituted C3-12 cycloalkyl, C4-20 cy cl oal
kyl alkyl, substituted
C4-20 cycloalkylalkyl, C4-20 heterocycloalkylalkyl, substituted C4-20
heterocycloalkylalkyl, C5.
12 aryl, substituted C5-12 aryl, C5-12 heteroaryl, substituted C5-12
heteroaryl, C6-20 arylalkyl,
substituted C6-20 arylalkyl, C6-20 heteroarylalkyl or substituted C6-20
heteroarylalkyl; and
n is an integer from 1 to 2.
[00124] In certain embodiments of a compound of Formula (VI), each R5, R1
and R22
is independently C1.6 alkyl, substituted C1.6 alkyl, C3.7 cycloalkyl,
substituted C3.7 cycloalkyl,
C5-7 aryl or substituted C5.7 aryl.
[00125] In certain embodiments of a compound of Formula (VI), each R5, R1
and R22
is independently hydrogen, methyl, ethyl, n-propyl, isopropyl, butyl,
isobutyl, sec-butyl, tert-
butyl, n-pentyl, isopentyl, sec-pentyl, neopentyl, dodecyl, 1,1 -
dimethoxyethyl, 1,1 -
diethoxyethyl, phenyl, 4-methoxyphenyl, benzyl, phenethyl, styryl,
cyclopropyl, cyclobutyl,
cyclopentyl, cyclohexyl, 2-pyridyl, 3 -pyridyl or 4-pyri dyl.
[00126] In certain embodiments of a compound of Formula (VI), each R5, R1
and R22
is independently hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl,
isobutyl, sec-butyl,
tert-butyl, n-pentyl, isopentyl, sec-pentyl, neopentyl, dodecyl, 1,1-
diethoxyethyl, phenyl,
cyclohexyl or 3-pyridyl.
[00127] In certain embodiments of a compound of Formula (VI), each R5, Rth
and R22
is independently hydrogen, methyl, ethyl, n-propyl, isopropyl, dodecyl, tert-
butyl, phenyl or
cyclohexyl
[00128] In certain embodiments of a compound of Formula (VI), each R5, R1
and R22
is independently ethyl, isopropyl or dodecyl.
[00129] In certain embodiments of a compound of Formula (VI), each le and
R4 is
independently hydrogen.
[00130] In certain embodiments of a compound of Formula (VI), R11 and R12
are each
hydroxyl
[00131] In certain embodiments of a compound of Formula (VI) one of R11 or
R12 is
hydrogen and the other is hydroxyl.
[00132] In certain embodiments of a compound of Formula (VI), each R23 is
hydrogen,
methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl, phenyl or cyclohexyl.
[00133] In certain embodiments of a compound of Formula (VI), each R23 is
methyl.
[00134] In certain embodiments of a compound of Formula (VI), each
substituent
group is independently halogen, -NO2, -OH, -NH7, -CN, -CF3, -0CF3, =0, C1-12
alkyl,
44

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
substituted C1-12 alkyl, C1-12 alkoxy or substituted C1-12 alkoxy, -COOR1 '
wherein is
hydrogen, C1-3 alkyl or -(NR11.)2 wherein each R1r is independently hydrogen
or C1.3 alkyl.
[00135] In certain embodiments of a compound of Formula (VI), n is the
integer 1.
[00136] In still another embodiment, the compound of Formula (VI) is a
compound of
Formula (XXII), Formula (XXIII), Formula (XXIV), Formula (XXV), Formula
(XXVI),
Formula (XXVII) or Formula (XXVIII) or a pharmaceutically acceptable salt,
solvate,
tautomer or stereoisomer thereof;
wherein the compound of formula (XXII) is:
HO ,=,õ
HNN /s/C) R32
RaR 0
(XXII)
wherein the compound of Formula (XXIII) is:
HO
HO H N N /o\ R32
0
wherein the compound of Formula (XXIV) is:
R33o
H2N N ./()'\ R32
0
(XXIV)
wherein the compound of Formula (XXV) is:

CA 02971729 2017-06-20
WO 2016/106284
PCT/US2015/067283
NH
HO HN N R32
0
(XXV)
wherein the compound of Formula (XXVI) is:
R 33o
R330.
HN N R32
0
(XXVI)
wherein the compound of Formula (XXVII) is:
HO.
R330
HN N R32
0
(XXVII)
wherein the compound of Formula (XXVIII) is:
R33
N
0
R33-1(\ R32
0
(XXVIII)
wherein R is ¨CH3 or ¨CD3;
le is hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-
butyl;
R32 is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl,
phenyl,
cyclohexyl;
46

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
R3 R4 R39
_c.2_c(0)0R43,_c.2_(o)c(o)R43,_c.2_(0)c(0)0R43 or 0;
R39 is hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl, dodecyl,
phenyl or
cyclohexyl;
each R33 is independently hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-
butyl,
dodecyl, phenyl or cyclohexyl; R43 is hydrogen, methyl, ethyl, n-propyl,
isopropyl, tert-butyl,
dodecyl, phenyl or cyclohexyl; and
R3 and R4 are each independently hydrogen, C1_12 alkyl or substituted C2
alkyl.
[00137] In certain embodiments each R32 and R33 is independently ethyl,
isopropyl or
dodecyl.
[00138] In certain embodiments R39 is methyl.
[00139] In certain embodiments R3 and R4 are each hydrogen.
[00140] In certain embodiments a creatine prodrug is a compound of Formula
(VII) or
a pharmaceutically acceptable salt, solvate, tautomer or stereoisomer thereof:
wherein the compound of Formula (VII) is:
R14
R14
o=¨=
jõ.
NH2
(VII)
wherein:
R is ¨CH3 or ¨CD3;
each R1-4 is independently hydrogen, C1-12 alkyl, substituted C1-12 alkyl, C1-
12
heteroalkyl, substituted C1-12 heteroalkyl, C3-12 cycloalkyl, substituted C3-
12 cycloalkyl, C4-20
cycloalkylalkyl, substituted C4-20 cycloalkylalkyl, C4-20
heterocycloalkylalkyl, substituted C4.
20 heterocycloalkylalkyl, C5-12 aryl, substituted Cu aryl, C.5.12 heteroaryl,
substituted C5-12
heteroaryl, C6_20 arylalkyl, substituted C6.70 arylalkyl, C6-20
heteroarylalkyl, substituted C6-20
heteroarylalkyl -CH(0R5), -C(0)R5, -C(0)0R5 or -C(0)(NR3R4);
each R3 and R4 is independently hydrogen, C142 alkyl or substituted C1.12
alkyl; and
47

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
R5 is hydrogen, Cm, alkyl, substituted C1-12 alkyl, C1-12 heteroalkyl,
substituted C1-12
heteroalkyl, C3-12 cycloalkyl, substituted C3-12 cycloalkyl, C4-20
cycloalkylalkyl, substituted
C4-20 cycloalkylalkyl, C4-20 heterocycloalkylalkyl, substituted C4-20
heterocycloalkylalkyl, C5.
12 aryl, substituted C5-12 aryl, C5-12 heteroaryl, substituted C5-12
heteroaryl, C6-20 arylalkyl,
substituted C6-20 arylalkyl, C6-20 heteroarylalkyl or substituted C6-20
heteroarylalkyl.
[00141] In certain embodiments of a compound of Formula (VII), R5 is C1.6
alkyl,
substituted C1-6 alkyl, C3.7 cycloalkyl, substituted C7 cycloalkyl, C5-7 aryl
or substituted C.7
aryl.
[00142] In certain embodiments of a compound of Formula (VII), R5 is
hydrogen,
methyl, ethyl, n-propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, n-
pentyl, isopentyl,
sec-pentyl, neopentyl, dodecyl, 1,1-dimethoxyethyl, 1,1-diethoxyethyl, phenyl,
4-
methoxyphenyl, benzyl, phenethyl, styryl, cyclopropyl, cyclobutyl,
cyclopentyl, cyclohexyl,
2-pyridyl, 3-pyridyl or 4-pyridyl
[00143] In certain embodiments of a compound of Formula (VII), R5 is
hydrogen,
methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl,
n-pentyl, isopentyl,
sec-pentyl, neopentyl, dodecyl, 1,1-diethoxyethyl, phenyl, cyclohexyl or 3-
pyridyl.
[00144] In certain embodiments of a compound of Formula (VII), R5 is
hydrogen,
methyl, ethyl, n-propyl, isopropyl, dodecyl, tert-butyl, phenyl or cyclohexyl.
[00145] In certain embodiments of a compound of Formula (VII), R5 is ethyl,
isopropyl or dodecyl.
[00146] In certain embodiments of a compound of Formula (VII), each R" is
independently hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-butyl,
dodecyl, phenyl or
cyclohexyl
[00147] In certain embodiments of a compound of Formula (VII), one R14 is
methyl
and the other R14 is hydrogen.
[00148] In certain embodiments of a compound of Formula (VII), each R3 and
R4 is
independently hydrogen
[00149] In certain embodiments of a compound of Formula (VII), each
substituent
group is independently halogen, -NO2, -OH, -NE17, -CN, -CF3, -0CF3, =0, C1.12
alkyl,
substituted C1-12 alkyl, C1-12 alkoxy or substituted C1-12 alkoxy, -COOR1 '
wherein RI . is
hydrogen, C1-3 alkyl or -(
NRi )2 wherein each Ril: is independently hydrogen or C1.3 alkyl.
[00150] In yet another embodiment, the compound of Formula (VII) is a
compound of
Formula (XXIX) or a pharmaceutically acceptable salt, solvate, tautomer or
stereoisomer
thereof;
48

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
wherein the compound of Formula (XXIX) is.
R34 R34
NH2
(XXIX)
wherein R is ¨CH3 or ¨CD3; and
each R34 is independently hydrogen, methyl, ethyl, n-propyl, isopropyl, tert-
butyl,
dodecyl, phenyl or cyclohexyl.
[00151] In certain embodiments of a compound of Formula (XXIX), one R34 is
methyl
and the other R34 is hydrogen.
Synthesis of Creatine Prodrugs
[00152] Those of ordinary skill in the art will appreciate that creatine
prodrug
compounds of Formulae (I), (III), (VI), (VII), and any subgenera or species
thereof, or a
pharmaceutically acceptable salt, solvate, tautomer or stereoisomer thereof
may be prepared
via general synthetic methods available in the art (e.g., Wuts and Greene,
"Protective Groups
in Organic Synthesis," John Wiley 8z Sons, 4th ed. 2006; Harrison et al.,
"Compendium of
Organic Synthetic Methods," Vols. 1-11, John Wiley 8z Sons 1971-2003; Larock
"Comprehensive Organic Transformations," John Wiley & Sons, 2nd ed. 2000; and
Paquette,
"Encyclopedia of Reagents for Organic Synthesis," John Wiley & Sons, 11th ed.
2003).
Starting materials useful for preparing compounds and intermediates thereof,
are
commercially available or can be prepared by well-known synthetic methods.
Pharmaceutical Compositions
[00153] Pharmaceutical compositions of the invention can comprise a
compound of the
invention and a pharmaceutically acceptable vehicle. A pharmaceutical
composition can
comprise a therapeutically effective amount of compound of the invention and a
pharmaceutically acceptable vehicle. In certain embodiments, a pharmaceutical
composition
can include more than one compound of the invention. Pharmaceutically
acceptable vehicles
include diluents, adjuvants, excipients, and carriers.
49

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
[00154] Pharmaceutical compositions can be produced using standard
procedures (see,
e.g., "Remington's The Science and Practice of Pharmacy," 21st edition,
Lippincott, Williams
& Wilcox, 2005). Pharmaceutical compositions may be manufactured by means of
conventional mixing, dissolving, granulating, dragee-making, levigating,
emulsifying,
encapsulating, entrapping or lyophilizing processes. Pharmaceutical
compositions may be
formulated in a conventional manner using one or more physiologically
acceptable carriers,
diluents, excipients or auxiliaries, which facilitate processing of compounds
disclosed herein
into preparations, which can be used pharmaceutically. Proper foimulation can
depend, in
part, on the route of administration
[00155] Pharmaceutical compositions of the invention can provide
therapeutic plasma
concentrations of a creatine creatine upon administration to a patient. The
promoiety of a
creatine prodrug can be cleaved in vivo either chemically and/or enzymatically
to release
creatine. One or more enzymes present in the intestinal lumen, intestinal
tissue, blood, liver,
brain or any other suitable tissue of a mammal can enzymatically cleave the
promoiety of the
administered prodrugs. For example, the promoiety can be cleaved after
absorption by the
gastrointestinal tract (e.g., in intestinal tissue, blood, liver or other
suitable tissue of a
mammal). In certain embodiments, a creatine remains conjugated to the
promoiety during
transit across the intestinal mucosal barrier to provide protection from
presystemic
metabolism. In certain embodiments, a creatine prodrug is essentially not
metabolized to
release the corresponding creatine within enterocytes, but is metabolized to
the parent drug
within the systemic circulation. Cleavage of the promoiety of a creatine
prodrug after
absorption by the gastrointestinal tract may allow the prodrug to be absorbed
into the
systemic circulation either by active transport, passive diffusion or by a
combination of both
active and passive processes.
[00156] Creatine prodrugs can remain intact until after passage of the
prodrug through
a biological barrier, such as the blood-brain barrier. In certain embodiments,
prodrugs of the
invention can be partially cleaved, e.g., one or more, but not all, of the
promoieties can be
cleaved before passage through a biological barrier or prior to being taken up
by a cell, tissue
or organ.
[00157] Creatine prodrugs can remain intact in the systemic circulation and
be
absorbed by cells of an organ, either passively or by active transport
mechanisms. In certain
embodiments, a creatine prodrug will be lipophilic and can passively
translocate through
cellular membranes. Following cellular uptake, the prodrug can be cleaved
chemically and/or
enzymatically to release the corresponding creatine into the cellular
cytoplasm, resulting in

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
an increase in the intracellular concentration of the creatine. In certain
embodiments, a
prodrug can be permeable to intracellular membranes such as the mitochondrial
membrane,
and thereby facilitate delivery of a prodrug, and following cleavage of the
promoiety or
promoieties, a creatine, to an intracellular organelle such as mitochondria.
[00158] In certain embodiments, a pharmaceutical composition can include an
adjuvant that facilitates absorption of a compound of the invention through
the
gastrointestinal epithelia. Such enhancers can, for example, open the tight-
junctions in the
gastrointestinal tract or modify the effect of cellular components, such as p-
glycoprotein and
the like. Suitable enhancers can include alkali metal salts of salicylic acid,
such as sodium
salicylate, caprylic or capric acid, such as sodium caprylate or sodium
caprate, and the like.
Enhancers can include, for example, bile salts, such as sodium deoxycholate.
Various p-
glycoprotein modulators are described in U.S. Pat. No. 5,112,817 and U.S. Pat.
No.
5,643,909. Various absorption enhancing compounds and materials are described
in U.S. Pat.
No. 5,824,638, and U.S. Application No. 2006/0046962. Other adjuvants that
enhance
permeability of cellular membranes include resorcinol, surfactants,
polyethylene glycol, and
bile acids.
[00159] In certain embodiments, a pharmaceutical composition can include an
adjuvant that reduces enzymatic degradation of a compound of of the invention.
Microencapsulation using protenoid microspheres, liposomes or polysaccharides
can also be
effective in reducing enzymatic degradation of administered compounds.
[00160] A pharmaceutical composition can also include one or more
pharmaceutically
acceptable vehicles, including excipients, adjuvants, carriers, diluents,
binders, lubricants,
disintegrants, colorants, stabilizers, surfactants, fillers, buffers,
thickeners, emulsifiers,
wetting agents, and the like. Vehicles can be selected to alter the porosity
and permeability of
a pharmaceutical composition, alter hydration and disintegration properties,
control
hydration, enhance manufacturability, etc.
[00161] In certain embodiments, a pharmaceutical composition can be
formulated for
oral administration. Pharmaceutical compositions formulated for oral
administration can
provide for uptake of a compound of the invention throughout the
gastrointestinal tract or in a
particular region or regions of the gastrointestinal tract. In certain
embodiments, a
pharmaceutical composition can be formulated to enhance uptake a compound of
the
invention from the upper gastrointestinal tract, and in certain embodiments,
from the small
intestine. Such compositions can be prepared in a manner known in the
pharmaceutical art
and can further comprise, in addition to a compound of the invention, one or
more
51

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
pharmaceutically acceptable vehicles, permeability enhancers, and/or a second
therapeutic
agent.
[00162] In certain embodiments, a pharmaceutical composition can further
comprise a
substance to enhance, modulate and/or control release, bioavailability,
therapeutic efficacy,
therapeutic potency, stability, and the like. For example, to enhance
therapeutic efficacy a
compound of the invention can be co-administered with one or more active
agents to increase
the absorption or diffusion of the drug from the gastrointestinal tract or to
inhibit degradation
of the drug in the systemic circulation. In certain embodiments, a compound of
the invention
can be co-administered with active agents having pharmacological effects that
enhance the
therapeutic efficacy of the compound of the invention.
[00163] In certain embodiments, a pharmaceutical composition can further
comprise
sub stances to enhance, modulate and/or control release, bi oavailability,
therapeutic efficacy,
therapeutic potency, stability, and the like. For example, to enhance
therapeutic efficacy a
compound of the invention can be co-administered with one or more active
agents to increase
the absorption or diffusion of a compound of the invention from the
gastrointestinal tract or
to inhibit degradation of the drug in the systemic circulation. In certain
embodiments, a
compound of the invention can be co-administered with active agents having
pharmacological effects that enhance the therapeutic efficacy of a compound of
the invention.
[00164] Pharmaceutical compositions can take the form of solutions,
suspensions,
emulsions, tablets, pills, pellets, capsules, capsules containing liquids,
powders, sustained-
release formulations, suppositories, emulsions, aerosols, sprays, suspensions
or any other
form suitable for use. Pharmaceutical compositions for oral delivery may be in
the form of
tablets, lozenges, aqueous or oily suspensions, granules, powders, emulsions,
capsules,
syrups or elixirs, for example. Orally administered compositions may contain
one or more
optional agents, for example, sweetening agents such as fructose, aspartame or
saccharin,
flavoring agents such as peppermint, oil of wintergreen or cherry coloring
agents and
preserving agents, to provide a pharmaceutically palatable preparation.
Moreover, when in
tablet or pill form, the compositions may be coated to delay disintegration
and absorption in
the gastrointestinal tract, thereby providing a sustained action over an
extended period of
time. Oral compositions can include standard vehicles such as mannitol,
lactose, starch,
magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc.
Such vehicles
can be of pharmaceutical grade. For oral liquid preparations such as, for
example,
suspensions, elixirs, and solutions, suitable carriers, excipients or diluents
include water,
saline, alkyleneglycols (e.g., propylene glycol), polyalkylene glycols (e.g.,
polyethylene
52

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
glycol) oils, alcohols, slightly acidic buffers between pH 4 and pH 6 (e.g.,
acetate, citrate,
ascorbate at between about 5 mM to about 50 mM), etc. Additionally, flavoring
agents,
preservatives, coloring agents, bile salts, acylcarnitines, and the like may
be added.
[00165] When a compound of the invention is acidic, it may be included in
any of the
above-described formulations as the free acid, a pharmaceutically acceptable
salt, solvate, a
solvate or a hydrate. Pharmaceutically acceptable salts substantially retain
the activity of the
free acid, may be prepared by reaction with bases, and tend to be more soluble
in aqueous
and other protic solvents than the corresponding free acid form. In some
embodiments,
sodium salts of a compound of the invention are used in the above-described
formulations.
[00166] Pharmaceutical compositions of the invention can formulated for
parenteral
administration including administration by injection, for example, into a vein
(intravenously),
an artery (intraarterially), a muscle (intramuscularly), under the skin
(subcutaneously or in a
depot formulation), to the pericardium, to the coronary arteries or used as a
solution for
delivery to a tissue or organ, for example, use in a cardiopulmonary bypass
machine or to
bathe transplant tissues or organs. Injectable compositions can be
pharmaceutical
compositions for any route of injectable administration, including, but not
limited to,
intravenous, intrarterial, intracoronary, pericardial, perivascular,
intramuscular, subcutaneous,
intradermal, intraperitoneal, and intraarticular. In certain embodiments, an
injectable
pharmaceutical composition can be a pharmaceutically appropriate composition
for
administration directly into the heart, pericardium or coronary arteries.
[00167] Pharmaceutical compositions of the invention suitable for
parenteral
administration can comprise one or more compounds of the invention in
combination with
one or more pharmaceutically acceptable sterile isotonic aqueous, water-
miscible or non-
aqueous vehicles. Pharmaceutical compositions for parenteral use may include
substances
that increase and maintain drug solubility such as complexing agents and
surface acting
agents, compounds that make the solution isotonic or near physiological pH
such as sodium
chloride, dextrose, and glycerin, substances that enhance the chemical
stability of a solution
such as antioxidants, inert gases, chelating agents, and buffers, substances
that enhance the
chemical and physical stability, substances that minimize self aggregation or
interfacial
induced aggregation, substances that minimize protein interaction with
interfaces,
preservatives including antimicrobial agents, suspending agents, emulsifying
agents, and
combinations of any of the foregoing. Pharmaceutical compositions for
parenteral
administration can be formulated as solutions, suspensions, emulsions,
liposomes,
microspheres, nanosystems, and powder to be reconstituted as solutions.
Parenteral
53

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
preparations are described in "Remington, The Science and Practice of
Pharmacy," 21st
edition, Lippincott, Williams & Wilkins, Chapter 41-42, pages 802-849, 2005.
[00168] In certain embodiments a pharmaceutical composition can be
formulated for
bathing transplantation tissue or organs before, during or after transit to an
intended recipient.
Such compositions can be used before or during preparation of a tissue or
organ for
transplant. In certain embodiments, a pharmaceutical composition can be a
cardioplegic
solution administered during cardiac surgery. In certain embodiments, a
pharmaceutical
composition can be used, for example, in conjunction with a cardiopulmonary
bypass
machine to provide the pharmaceutical composition to the heart. Such
pharmaceutical
compositions can be used during the induction, maintenance or reperfusion
stages of cardiac
surgery (see e.g., Chang et al., Masui 2003, 52(4), 356-62; Ibrahim et al.,
Eur. J. Cardiothorac
Surg 1999, 15(1), 75-83; von Oppell et al., J Thorac Cardiovasc Surg. 1991,
102(3), 405-12;
and Ji et al., J. Extra Corpor Technol 2002, 34(2), 107-10). In certain
embodiments, a
pharmaceutical composition can be delivered via a mechanical device such as a
pump or
perfuser (see e.g., Hou and March, J Invasive Cardiol 2003, 15(1), 13-7;
Maisch et al., Am. J
Cardiol 2001, 88(11), 1323-6; and Macris and Igo, Clin Cardiol 1999, 22 (1,
Suppl 1), 136-
9).
[00169] For prolonged delivery, a pharmaceutical composition can be
provided as a
depot preparation, for administration by implantation, e.g., subcutaneous,
intradermal or
intramuscular injection. Thus, in certain embodiments, a pharmaceutical
composition can be
formulated with suitable polymeric or hydrophobic materials, e.g., as an
emulsion in a
pharmaceutically acceptable oil, ion exchange resins or as a sparingly soluble
derivative, e.g.,
as a sparingly soluble salt form of a compound of the invention.
[00170] Pharmaceutical compositions of the invention can be formulated so
as to
provide immediate, sustained or delayed release of a compound of Formula (1)
and/or
Formula (II) after administration to the patient by employing procedures known
in the art
(see, e.g., Allen et al., "Ansel's Pharmaceutical Dosage Forms and Drug
Delivery Systems,"
8th ed., Lippincott, Williams & Wilkins, August 2004).
Dosage Forms
[00171] Pharmaceutical compositions of the invention can be foimulated in a
unit
dosage form. Unit dosage form refers to a physically discrete unit suitable as
a unitary dose
for patients undergoing treatment, with each unit containing a predetermined
quantity of a
compound of the invention calculated to produce an intended therapeutic
effect. A unit
54

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
dosage foun can be for a single daily dose or one of multiple daily doses,
e.g., 2 to 4 times
per day. When multiple daily doses are used, the unit dosage can be the same
or different for
each dose. One or more dosage forms can comprise a dose, which may be
administered to a
patient at a single point in time or during a time interval.
[00172] Pharmaceutical compositions of the invention can be used in dosage
forms
that provide immediate release and/or controlled release of a compound of the
invention. The
appropriate type of dosage form can depend on the disease, disorder or
condition being
treated, and on the method of administration. For example, for the treatment
of acute
ischemic conditions such as cardiac failure or stroke the use of an immediate
release
pharmaceutical composition or dosage form administered parenterally may be
appropriate.
For treatment of chronic neurodegenerative disorders, controlled release
pharmaceutical
composition or dosage form administered orally may be appropriate.
[00173] In certain embodiments, a dosage form can be adapted to be
administered to a
patient no more than twice per day, and in certain embodiments, only once per
day. Dosing
may be provided alone or in combination with other drugs and may continue as
long as
required for effective treatment of the disease, disorder or condition.
[00174] Pharmaceutical compositions comprising a compound of the invention
can be
formulated for immediate release for parenteral administration oral
administration or by any
other appropriate route of administration.
[00175] Controlled drug delivery systems can be designed to deliver a drug
in such a
way that the drug level is maintained within the therapeutic windows and
effective and safe
blood levels are maintained for a period as long as the system continues to
deliver the drug at
a particular rate. Controlled drug delivery can produce substantially constant
blood levels of a
drug as compared to fluctuations observed with immediate release dosage forms.
For some
drugs, maintaining a constant bloodstream and tissue concentration throughout
the course of
therapy is the most desirable mode of treatment. Immediate release of these
drugs can cause
blood levels to peak above the level required to elicit the desired response,
which wastes the
drug and may cause or exacerbate toxic side effects. Controlled drug delivery
can result in
optimum therapy, and not only can reduce the frequency of dosing, and may also
reduce the
severity of side effects. Examples of controlled release dosage forms include
dissolution
controlled systems, diffusion controlled systems, ion exchange resins,
osmotically controlled
systems, erodable matrix systems, pH independent formulations, gastric
retention systems,
and the like.

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
[00176] In certain embodiments, an oral dosage form of the invention can be
a
controlled release dosage foini. Controlled delivery technologies can improve
the absorption
of a drug in a particular region or regions of the gastrointestinal tract. The
appropriate oral
dosage form for a particular pharmaceutical composition of the invention can
depend, at least
in part, on the gastrointestinal absorption properties of the compound of the
invention, the
stability of the compound of the invention in the gastrointestinal tract, the
pharmacokinetics
of the compound of the invention, and the intended therapeutic profile. An
appropriate
controlled release oral dosage form can be selected for a particular the
compound of the
invention. For example, gastric retention oral dosage forms can be appropriate
for compounds
absorbed primarily from the upper gastrointestinal tract, and sustained
release oral dosage
forms can be appropriate for compounds absorbed primarily form the lower
gastrointestinal
tract.
[00177] Certain compounds are absorbed primarily from the small intestine.
In general,
compounds traverse the length of the small intestine in about 3 to 5 hours.
For compounds
that are not easily absorbed by the small intestine or that do not dissolve
readily, the window
for active agent absorption in the small intestine may be too short to provide
a desired
therapeutic effect. Gastric retention dosage forms, i.e., dosage forms that
are designed to be
retained in the stomach for a prolonged period of time, can increase the
bioavailability of
drugs that are most readily absorbed by the upper gastrointestinal tract. The
residence time of
a conventional dosage form in the stomach is 1 to 3 hours. After transiting
the stomach, there
is approximately a 3 to 5 hour window of bioavailability before the dosage
form reaches the
colon. However, if the dosage form is retained in the stomach, the drug can be
released
before it reaches the small intestine and will enter the intestine in solution
in a state in which
it can be more readily absorbed. Another use of gastric retention dosage forms
is to improve
the bioavailability of a drug that is unstable to the basic conditions of the
intestine (see, e.g.,
Hwang et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1998, 15,
243-284) To
enhance drug absorption from the upper gastrointestinal tract, several gastric
retention dosage
forms have been developed. Examples include, hydrogels (see, e.g., U.S.
Application No.
2003/0008007), buoyant matrices (see, e.g., U.S. Application No.
2006/0013876), polymer
sheets (see, e.g., U.S. Application No. 2005/0249798), microcellular foams
(see, e.g., U.S.
Application No. 2005/0202090), and swellable dosage forms (see, e.g., U.S.
Application No.
2005/0019409; U.S. Pat. No. 6,797,283; U.S. Application No. 2006/0045865; U.S.
Application No. 2004/0219186; U.S. Pat. No. 6,723,340; U.S. Pat. No.
6,476,006; U.S. Pat.
No. 6,120,803; U.S. Pat. No. 6,548,083; U.S. Pat. No. 6,635,280; U.S. Pat. No.
5,780,057).
56

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
Bioadhesive polymers can also provide a vehicle for controlled delivery of
drugs to a number
of mucosal surfaces in addition to the gastric mucosa (see, e.g., U.S. Pat.
No. 6,235,313; U.S.
Pat. No. 6,207,197; U.S. Application No. 2006/0045865 and U.S. Application No.
2005/0064027). Ion exchange resins have been shown to prolong gastric
retention, potentially
by adhesion.
[00178] In a swelling and expanding system, dosage forms that swell and
change
density in relation to the surrounding gastric content can be retained in the
stomach for longer
than a conventional dosage form. A dosage form can absorb water and swell to
fon') a
gelatinous outside surface and float on the surface of gastric content surface
while
maintaining integrity before releasing a drug. Fatty materials can be added to
impede wetting
and enhance flotation when hydration and swelling alone are insufficient.
Materials that
release gases may also be incorporated to reduce the density of a gastric
retention dosage
form. Swelling also can significantly increase the size of a dosage form and
thereby impede
discharge of the non-disintegrated swollen solid dosage form through the
pylorus into the
small intestine. Swellable dosage forms can be formed by encapsulating a core
containing
drug and a swelling agent or by combining a drug, swelling agent, and one or
more erodible
polymers.
[00179] Gastric retention dosage forms can also be in the form of a folded
thin sheet
containing a drug and water-insoluble diffusible polymer that opens in the
stomach to its
original size and shape, which is sufficiently large to prevent or inhibit
passage of the
expanded dosage from through the pyloric sphincter.
[00180] Floating and buoyancy gastric retention dosage forms can be
designed to trap
gases within sealed encapsulated cores that can float on the gastric contents,
and thereby be
retained in the stomach for a longer time, e.g., 9 to 12 hours. Due to the
buoyancy effect,
these systems can provide a protective layer preventing the reflux of gastric
content into the
esophageal region and can also be used for controlled release devices. A
floating system can,
for example, contain hollow cores containing drug coated with a protective
membrane. The
trapped air in the cores floats the dosage from on the gastric content until
the soluble
ingredients are released and the system collapses. In other floating systems,
cores contain
drug and chemical substances capable of generating gases when activated. For
example,
coated cores, containing carbonate and/or bicarbonate can generate carbon
dioxide in the
reaction with hydrochloric acid in the stomach or incorporated organic acid in
the system.
The gas generated by the reaction is retained to float the dosage form. The
inflated dosage
57

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
form later collapses and clears form the stomach when the generated gas
permeates slowly
through the protective coating.
[00181] Bioadhesive polymers can also provide a vehicle for controlled
delivery of
drugs to a number of mucosal surfaces in addition to the gastric mucosa (see,
e.g., U.S. Pat.
No. 6,235,313; and U.S. Pat. No. 6,207,197). A bioadhesive system can be
designed by
incorporation of a drug and other excipients within a bioadhesive polymer. On
ingestion, the
polymer hydrates and adheres to the mucus membrane of the gastrointestinal
tract.
Bioadhesive polymers can be selected that adhere to a desired region or
regions of the
gastrointestinal tract. Bioadhesive polymers can be selected to optimized
delivery to targeted
regions of the gastrointestinal tract including the stomach and small
intestine. The mechanism
of the adhesion is thought to be through the formation of electrostatic and
hydrogen bonding
at the polymer-mucus boundary. U.S. Application Nos. 2006/0045865 and
2005/0064027
disclose bioadhesive delivery systems which are useful for drug delivery to
both the upper
and lower gastrointestinal tract.
[00182] Ion exchange resins have been shown to prolong gastric retention,
potentially
by adhesion.
[00183] Gastric retention oral dosage forms can be appropriately used for
delivery of
drugs that are absorbed mainly from the upper gastrointestinal tract. For
example, certain
compounds of the invention may exhibit limited colonic absorption, and be
absorbed
primarily from the upper gastrointestinal tract. Thus, dosage forms that
release the compound
of the invention in the upper gastrointestinal tract and/or retard transit of
the dosage form
through the upper gastrointestinal tract will tend to enhance the oral
bioavailability of the
compound of the invention. Other forms of creatine prodrugs disclosed herein
can be
appropriately used with gastric retention dosage forms.
[00184] Polymer matrices have also been used to achieve controlled release
of the drug
over a prolonged period of time. Such sustained or controlled release can be
achieved by
limiting the rate by which the surrounding gastric fluid can diffuse through
the matrix and
reach the drug, dissolve the drug and diffuse out again with the dissolved
drug or by using a
matrix that slowly erodes, continuously exposing fresh drug to the surrounding
fluid.
Disclosures of polymer matrices that function by these methods are found, for
example, in
Skinner, U.S. Pat. Nos. 6,210,710 and 6,217,903; U.S. Pat. No. 5,451,409; U.S.
Pat. No.
5,945,125; PCT International Publication No. WO 96/26718; U.S. Pat. No.
4,915,952; U.S.
Pat. No. 5,328,942; U.S. Pat. No. 5,783,212; U.S. Pat. No. 6,120,803; and U.S.
Pat. No.
6,090,411.
58

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
[00185] Other drug delivery devices that remain in the stomach for extended
periods of
time include, for example, hydrogel reservoirs containing particles (U.S. Pat.
No. 4,871,548);
swellable hydroxypropylmethylcellulose polymers (U.S. Pat. No. 4,871,548);
planar
bioerodible polymers (U.S. Pat. No. 4,767,627); plurality of compressible
retention arms
(U.S. Pat. No. 5,443,843); hydrophilic water-swellable, cross-linked polymer
particles (U.S.
Pat. No. 5,007,790); and albumin-cross-linked polyvinylpyrrolidone hydrogels
(Park et al., J.
Controlled Release 1992, 19, 131-134).
[00186] In certain embodiments, pharmaceutical compositions of the
invention can be
practiced with a number of different dosage forms, which can be adapted to
provide sustained
release of the compound of the invention upon oral administration. Sustained
release oral
dosage forms can be used to release drugs over a prolonged time period and are
useful when
it is desired that a drug or drug form be delivered to the lower
gastrointestinal tract Sustained
release oral dosage forms include diffusion-controlled systems such as
reservoir devices and
matrix devices, dissolution-controlled systems, osmotic systems, and erosion-
controlled
systems. Sustained release oral dosage forms and methods of preparing the same
are well
known in the art (see, for example, "Remington's Pharmaceutical Sciences,"
Lippincott,
Williams & Wilkins, 21st edition, 2005, Chapters 46 and 47; Langer, Science
1990, 249,
1527-1533; and Rosoff, "Controlled Release of Drugs," 1989, Chapter 2).
[00187] Sustained release oral dosage forms include any oral dosage form
that
maintains therapeutic concentrations of a drug in a biological fluid such as
the plasma, blood,
cerebrospinal fluid or in a tissue or organ for a prolonged time period.
Sustained release oral
dosage forms include diffusion-controlled systems such as reservoir devices
and matrix
devices, dissolution-controlled systems, osmotic systems, and erosion-
controlled systems.
Sustained release oral dosage forms and methods of preparing the same are well
known in the
art (see, for example, "Remington's: The Science and Practice of Pharmacy,"
Lippincott,
Williams & Wilkins, 21st edition, 2005, Chapters 46 and 47; Langer, Science
1990, 249,
1527-1533; and Rosoff, "Controlled Release of Drugs," 1989, Chapter 2).
[00188] In diffusion-controlled systems, a water-insoluble polymer controls
the flow of
fluid and the subsequent egress of dissolved drug from the dosage form. Both
diffusional and
dissolution processes are involved in release of drug from the dosage form. In
reservoir
devices, a core comprising a drug is coated with the polymer, and in matrix
systems, the drug
is dispersed throughout the matrix. Cellulose polymers such as ethylcellulose
or cellulose
acetate can be used in reservoir devices. Examples of materials useful in
matrix systems
include methacrylates, acrylates, polyethylene, acrylic acid copolymers,
polyvinylchloride,
59

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
high molecular weight polyvinylalcohols, cellulose derivates, and fatty
compounds such as
fatty acids, glycerides, and carnauba wax.
[00189] In
dissolution-controlled systems, the rate of dissolution of the drug is
controlled by slowly soluble polymers or by microencapsulation. Once the
coating is
dissolved, the drug becomes available for dissolution. By varying the
thickness and/or the
composition of the coating or coatings, the rate of drug release can be
controlled. In some
dissolution-controlled systems, a fraction of the total dose can comprise an
immediate-release
component. Dissolution-controlled systems include encapsulated/reservoir
dissolution
systems and matrix dissolution systems. Encapsulated dissolution systems can
be prepared by
coating particles or granules of drug with slowly soluble polymers of
different thickness or by
microencapsulation. Examples of coating materials useful in dissolution-
controlled systems
include gelatin, carnauba wax, shellac, cellulose acetate phthalate, and
cellulose acetate
butyrate. Matrix dissolution devices can be prepared, for example, by
compressing a drug
with a slowly soluble polymer carrier into a tablet form.
[00190] The rate
of release of drug from osmotic pump systems is determined by the
inflow of fluid across a semipermeable membrane into a reservoir, which
contains an osmotic
agent. The drug is either mixed with the agent or is located in a reservoir.
The dosage form
contains one or more small orifices from which dissolved drug is pumped at a
rate
determined by the rate of entrance of water due to osmotic pressure. As
osmotic pressure
within the dosage form increases, the drug is released through the orifice(s).
The rate of
release is constant and can be controlled within tight limits yielding
relatively constant
plasma and/or blood concentrations of the drug. Osmotic pump systems can
provide a
constant release of drug independent of the environment of the
gastrointestinal tract. The rate
of drug release can be modified by altering the osmotic agent and the sizes of
the one or more
orifices.
[00191] The
release of drug from erosion-controlled systems is determined by the
erosion rate of a carrier matrix. Drug is dispersed throughout the polymer and
the rate of drug
release depends on the erosion rate of the polymer. The drug-containing
polymer can degrade
from the bulk and/or from the surface of the dosage form.
[00192]
Sustained release oral dosage forms can be in any appropriate form for oral
administration, such as, for example, in the forni of tablets, pills or
granules. Granules can be
filled into capsules, compressed into tablets or included in a liquid
suspension. Sustained
release oral dosage forms can additionally include an exterior coating to
provide, for
example, acid protection, ease of swallowing, flavor, identification, and the
like.

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
[00193] In certain embodiments, sustained release oral dosage folins can
comprise a
therapeutically effective amount of a compound of the invention and a
pharmaceutically
acceptable vehicle. In certain embodiments, a sustained release oral dosage
form can
comprise less than a therapeutically effective amount of a compound of the
invention and a
pharmaceutically effective vehicle. Multiple sustained release oral dosage
foams, each dosage
form comprising less than a therapeutically effective amount of a compound of
the invention,
can be administered at a single time or over a period of time to provide a
therapeutically
effective dose or regimen for treating a disease in a patient associated with
a dysfunction in
energy metabolism such as, for example, ischemia, oxidative stress, a
neurodegenerative
disease, including amyotrophic lateral sclerosis (ALS), Huntington's disease,
Parkinson's
disease or Alzheimer's disease, ischemic reperfusion injury, a cardiovascular
disease,
multiple sclerosis (MS), a psychotic disorder, a genetic disease affecting the
creatine kinase
system or muscle fatigue
[00194] Sustained release oral dosage forms of the invention can release a
compound
of the invention from the dosage form to facilitate the ability of the
compound of the
invention to be absorbed from an appropriate region of the gastrointestinal
tract, for example,
in the small intestine or in the colon. In certain embodiments, a sustained
release oral dosage
from can release a compound of the invention from the dosage form over a
period of at least
about 4 hours, at least about 8 hours, at least about 12 hours, at least about
16 hours, at least
about 20 hours, and in certain embodiments, at least about 24 hours. In
certain embodiments,
a sustained release oral dosage form can release a compound of the invention
from the dosage
form in a delivery pattern of from about 0 wt % to about 20 wt % in about 0 to
about 4 hours,
about 20 wt % to about 50 wt ,/0 in about 0 to about 8 hours, about 55 wt %
to about 85 wt %
in about 0 to about 14 hours, and about 80 wt % to about 100 wt % in about 0
to about 24
hours. In certain embodiments, a sustained release oral dosage form can
release a compound
of Formula (I) and/or Formula (II) from the dosage form in a delivery pattern
of from about 0
wt % to about 20 wt % in about 0 to about 4 hours, about 20 wt % to about 50
wt % in about
0 to about 8 hours, about 55 wt % to about 85 wt % in about 0 to about 14
hours, and about
80 wt % to about 100 wt % in about 0 to about 20 hours. In certain
embodiments, a sustained
release oral dosage form can release a compound of the invention from the
dosage form in a
delivery pattern of from about 0 wt % to about 20 wt % in about 0 to about 2
hours, about 20
wt % to about 50 wt % in about 0 to about 4 hours, about 55 wt % to about 85
wt % in about
0 to about 7 hours, and about 80 wt % to about 100 wt % in about 0 to about 8
hours.
61

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
[00195] Sustained release oral dosage forms comprising a creatine prodrug
compound
of the invention can provide a concentration of creatine in the plasma, blood
or tissue of a
patient over time, following oral administration to the patient. The
concentration profile of
creatine can exhibit an AUC that is proportional to the dose of the
corresponding compound
of the invention.
[00196] Regardless of the specific form of controlled release oral dosage
foiin used, a
compound of the invention can be released from an orally administered dosage
form over a
sufficient period of time to provide prolonged therapeutic concentrations of
the compound of
the invention in the plasma and/or blood of a patient. Following oral
administration, a dosage
form comprising a compound of the invention can provide a therapeutically
effective
concentration of creatine in the plasma and/or blood of a patient for a
continuous time period
of at least about 4 hours, of at least about 8 hours, for at least about 12
hours, for at least
about 16 hours, and in certain embodiments, for at least about 20 hours
following oral
administration of the dosage form to the patient. The continuous time periods
during which a
therapeutically effective concentration of creatine is maintained can be the
same or different.
The continuous period of time during which a therapeutically effective plasma
concentration
of creatine is maintained can begin shortly after oral administration or after
a time interval.
[00197] In certain embodiments, an oral dosage for treating a disease,
disorder or
condition in a patient can comprise a compound of the invention wherein the
oral dosage
form is adapted to provide, after a single administration of the oral dosage
form to the patient,
a therapeutically effective concentration of creatine in the plasma of the
patient for a first
continuous time period selected from at least about 4 hours, at least about 8
hours, at least
about 12 hours, and at least about 16 hours, and at least about 20 hours.
Methods of Use
[00198] The creatine kinase (creatine-creatine phosphate) system serves a
number of
functions in maintaining intracellular energy homeostasis (see e.g., Walsh et
al., J Physiol,
2001, 537, 971-978). Phosphocreatine acts as a temporal energy buffer at
intracellular sites of
high energy translocation which operates when the rate of ATP utilization is
greater than the
rate of ATP production by mitochondrial respiration. Mitochondrial creatine
kinase allows
the high energy phosphate bond of newly synthesized ATP to be transferred to
creatine, thus
generating phosphocreatine, which is much more stable than ATP.
Phosphocreatine can
diffuse throughout a cell and its high energy phosphate bond can be used to
regenerate ATP
from ADP at heavy energy utilization sites where other creatine kinase enzymes
are
62

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
strategically positioned. These sites include membranes that engage in ion
transport, axonal
regions involved in transporting material along microtubules to and from
presynaptic
endings, and presynaptic endings where energy is required for
neurotransmission. Neurons
synthesize creatine, however the amount of creatine can be severely depleted
during injury.
As with skeletal and heart muscle, neuronal creatine stores can to some extent
be increased
by oral supplementation of creatine. The creatine kinase system also serves as
an intracellular
spatial energy transport mechanism. In this role as an energy carrier, energy
generated by the
ATP-ADP system within mitochondria is coupled to the creatine-creatine
phosphate system
in the cytosol, which in turn is coupled to extra-mitochondrial ATP-ADP
systems at sites of
high intracellular energy transduction. The creatine-creatine phosphate system
is also
believed to act as a low threshold ADP sensor that maintains ATP-ADP
concentration ratios
in subcellular locations wherein creatine kinase is functionally coupled to
ATP-consuming
and ATP-producing pathways. For example, it has been shown that creatine can
react with
ATP derived from mitochondrial respiration in a reaction catalyzed by
mitochondrial creatine
kinase and functionally coupled to adenine nucleotide translocase, thereby
resulting in an
increase in local ADP concentration and the stimulation of mitochondrial
respiration. The
creatine kinase system is therefore particularly important in maintaining
energy homeostasis,
including ATP homeostasis, in cells, tissues, and organs with high-energy
consumption
requirements such as neurons and muscles.
[00199] Compounds of the invention and pharmaceutical compositions of the
invention can be useful in treating of diseases, disorders or conditions in a
patient associated
with a dysfunction in energy metabolism. In certain embodiments, the
dysfunction in energy
metabolism comprises a depletion in intracellular ATP concentration, a
decreased
intracellular creatine phosphate concentration, a decreased intracellular
creatine phosphate to
ATP concentration ratio, and/or a dysfunction in the creatine kinase system in
a tissue or
organ affected by the disease. In certain embodiments, a dysfunction in energy
metabolism
comprises a decreased intracellular ATP concentration in a tissue or organ
affected by the
disease. In certain embodiments, a dysfunction in energy metabolism comprises
a decreased
intracellular creatine phosphate concentration in a tissue or organ affected
by the disease. In
certain embodiments, the dysfunction in energy metabolism comprises a
dysfunction in the
creatine kinase system and/or other intracellular energy pathway in a tissue
or organ affected
by the disease. In certain embodiments, a disease associated with a
dysfunction in energy
metabolism is selected from ischemia, oxidative stress, a neurodegenerative
disease, ischemic
reperfusion injury, a cardiovascular disease, multiple sclerosis, a psychotic
disease, and
63

muscle fatigue. In certain embodiments, treating a disease comprises effecting
energy homeostasis in a
tissue or organ affected by the disease.
[00200] Compounds of the invention and pharmaceutical compositions thereof
can be used to treat
a disease in a patient associated with oxidative stress by administering to a
patient in need of such
treatment a therapeutically effective amount of a compound of the invention or
a pharmaceutical
composition thereof. In certain embodiments, the oxidative stress is
associated with ischemia or a
neurodegenerative disorder. Methods of the invention include treating an
oxidatively stressed tissue or
organ by contacting the tissue or organ with a compound of the invention or a
pharmaceutical
composition thereof.
[00201] Compounds and pharmaceutical compositions of the invention can be
useful in treating
diseases, disorders or conditions in which a rapid increase in intracellular
creatine levels has a therapeutic
effect.
Ischemia
[00202] Compounds and pharmaceutical compositions of the invention can be
used to treat acute
or chronic ischemic diseases, disorders or conditions. Ischemia is an
imbalance of oxygen supply and
demand in a cell, tissue or organ. Ischemia is characterized by hypoxia,
including anoxia, insufficiency
of metabolic substrates for normal cellular bioenergetics, and accumulation of
metabolic waste. Ischemia
in a tissue or organ may be caused by a vascular insufficiency such as
arteriosclerosis, thrombosis,
embolism, torsion or compression, hypotension such as shock or hemorrhage,
increased tissue mass
(hypertrophy), increased workload (tachycardia, exercise), and/or by decreased
tissue stress such as
cardiac dilation. Ischemia can also result from trauma or surgical procedures.
Depending on the severity
and duration of the injury, ischemia can lead to a reversible loss of cellular
function or to irreversible cell
death. Different cell types have different thresholds to ischemic injury
depending, at least in part, on the
cellular energy requirements of the tissue(s) or organ(s) affected.
Parenchymal cells such as neurons (3-
4 minutes), cardiac muscles, hepatocytes, renal tubular cells,
gastrointestinal epithelium (20-80 minutes)
and fibroblasts, epidermis, and skeletal muscle (hours) are more susceptible
to ischemic injury than are
stromal cells. A number of studies suggest a correlation between the
functional capacity of the creatine
kinase system and ischemic tolerance of a given tissue, and indicate that
strategies toward improving the
functional capacity of the creatine kinase system may be effective for
improving ischemic tolerance in
tissue (see e.g., Wyss and Kaddurah-Daouk, Physiological Reviews, 2000, 80(3),
1107-1213). For
example oral
64
Date Recue/Date Received 2022-04-14

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
creatine supplementation inhibits mitochondrial cytochrome C release and
downstream
caspase-3 activation, resulting in ischemic neuroprotection. Associated with
inhibition of
cytochrome C release and caspase-3 activation and with neuroprotection,
creatine
administration inhibits ischemia-mediated ATP depletion.
[00203] Compounds and pharmaceutical compositions of the invention can be
used to
treat acute or chronic ischemia. In certain embodiments, a compound or
composition can be
particularly useful in acute or emergency treatment of ischemia in tissue or
organs
characterized by high energy demand such as the brain, neurons, heart, lung,
kidney or the
intestine.
[00204] The high energy requirements compared to the low energy reserves
render the
brain particularly vulnerable to hypoxic conditions. Although the brain
constitutes only a
small fraction of total body weight (about 2%), it accounts for a
disproportionately large
percentage of 02 consumption (about 20%). Under physiological conditions,
enhanced
demand for 02 is rapidly and adequately compensated for by an increase in
cerebral blood
flow. The longer the duration of hypoxia/ischemia, the larger and more diffuse
the areas of
the brain that are affected. The areas most vulnerable to ischemic damage are
the brainstem,
hippocampus, and cerebral cortex. Injury progresses and eventually becomes
irreversible
except if oxygenation is not restored. Acute cell death occurs mainly through
necrosis but
hypoxia also causes delayed apoptosis. In addition glutamate release from
presynaptic
neurons can further enhance Ca2+ influx and result in catastrophic collapse in
postsynaptic
cells. If the ischemia is not too severe, cells can suppress some functions,
i.e., protein
synthesis and spontaneous electrical activity, in a process called penumbra,
which can be
reversed provided that 02 supply is resumed. However, the process of restoring
oxygen levels
to ischemically stressed tissue, e.g., reperfusion, can also induce
irreversible cell death,
mainly through the generation of reactive oxygen species and inflammatory cell
infiltration.
[00205] The neuron is limited by its availability of energy-generating
substrates, being
limited to using primarily glucose, ketone bodies or lactate The neuron does
not produce or
store glucose or ketone bodies and cannot survive for any significant period
of time without a
substrate, which is absorbed and used directly or indirectly from the
bloodstream. Thus, a
constant supply of an energy-generating substrate must be present in the blood
at all times in
an amount sufficient to supply the entire brain and the rest of the body with
energy-
generating substrates. Brain cells require a concentration of about 5 mM
glucose (or its
equivalent) in order to maintain its optimal rate of oxidative phosphorylation
to produce ATP.
Nutrients enter cells by passing through the cell membrane. Nutrient delivery
frequently

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
relies upon mechanisms outside the cell membranes such as oral intake,
absorption,
circulatory transport and interstitial flux. Once localized in the vicinity of
the cell, membrane-
specific processes play a role in nutrient transport sequentially across the
blood-brain barrier
and then into the interior of the cell and into various subcellular
organelles. Nutrient transport
is made possible by the breakdown of ATP by ATPases. Na+ gradients created by
Na+/K+
ATPases can be used by cells to transport nutrient molecules across cell
membranes.
[00206] Lack of oxygen or glucose prevents or limits the ability of neurons
to
synthesize ATP. The intracellular creatine/phosphocreatine system can to some
extent
compensate for the lack of oxygen or glucose. Creatine kinase catalyses the
synthesis of
phosphocreatine from creatine in normal brain tissue. Under conditions of ATP
depletion,
phosphocreatine can donate its phosphate group to ADP to resynthesize ATP.
However,
neuronal phosphocreatine content is limited following complete anoxia or
ischemia
phosphocreatine is also rapidly depleted. ATP depletion is believed to block
Na+/K+ ATPases
causing neurons to depolarize and lose membrane potential.
[00207] Depleted oxygen levels have several other consequences on cellular
bioenergetics and function that can ultimately lead to cell death. For
example, dysfunctional
bioenergetics also involves impaired calcium homeostasis. The regulation of
calcium plays a
central role in the proper functioning and survival of neurons. Calcium pumps,
located on cell
membranes, use ATP to transport calcium ions out of the neuron. Proper
activity of the
calcium pump is essential in the maintenance of neuronal, mitochondrial, and
endoplasmic
reticulum homeostasis. Alterations in calcium pump function modulate enzyme
activity
within a cell and also play a critical role in triggering the mitochondrial
permeability
transition, which may lead to cell death. For example, intracellular Ca2+
metabolism is
believed to contribute to cell death in Alzheimer's disease. For example,
under conditions of
oxidative stress, the production of oxygen free radicals exceeds endogenous
free radical
protective mechanisms. This impairs neuronal metabolism and function by direct
free radical
damage to important cellular biomolecules including membrane lipids, nucleic
acids, and
functional proteins; and by modulation of critical signal transduction
pathways. Neural
function is dependent upon transmission of electrical impulses between cells.
This activity
relies upon the precise actions of multiple membrane proteins each suspended
in a
phospholipid bilayer. The optimal activity of this dynamic membrane
microenvironment
depends upon the exact status and chemical composition of the lipid
constituents. Lacking the
appropriate phospholipid environment, cell channel proteins, enzymes, and
receptors are not
able to achieve sustained levels of optimal function. In addition, oxidative
stress and/or
66

abnormal methyl metabolism can reduce the fluidity of the membranous lipid
bilayer with subsequent
adverse effects upon embedded functional proteins. Dysfunctional bioenergetics
may also adversely
affect passage of high-energy electrons along the respiratory chain.
[00208] Apoptosis refers to the energy-requiring process of programmed cell
death whereupon an
individual nerve cell under appropriate circumstances initiates a process
leading to cell death. Certain of
the mechanisms discussed above may initiate apoptotic pathways including
oxidative stress, calcium
overload, cellular energy deficiency, trophic factor withdrawal, and abnormal
amyloid precursor protein
processing. In ischemia, neurons in the brain tissue region that are most
severely affected by hypoxic
injury die rapidly by necrosis, whereas neurons exposed to lesser degrees of
hypoxia die by apoptosis.
The shift from necrotic cell death to apoptotic cell death is associated with
increasing levels of intra
cellular ATP. It has been shown that creatine supplementation can result in a
greater ability to buffer
ATP levels and reduce cell death and thereby provide protection from anoxic
and ischemic damage
(Balestrino et al., Amino Acids, 2002, 23, 221-229; and Zhu et al., J Neurosci
2004, 24(26), 5909-5912).
[00209] In certain embodiments, compounds and pharmaceutical compositions
of the invention
can be used to treat a cardiovascular disease, including cerebral ischemia
(stroke) and myocardial
ischemia (heart infarction). Ischemic heart disease, as the underlying cause
of many cases of acute
myocardial infarction, congestive heart failure, arrhythmias, and sudden
cardiac death, is a leading cause
of morbidity and mortality in all industrialized nations. In the United
States, ischemic heart disease causes
nearly 20% of all deaths (.about.600,000 deaths each year) with many of these
deaths occurring before
the patient arrives at the hospital. An estimated 1.1 million Americans will
have a new or recurrent acute
myocardial infarction each year, and many survivors will experience lasting
morbidity, with progression
to heart failure and death. As the population grows older and co-morbidities
such as obesity and diabetes
become more prevalent, the public health burden caused by ischemic heart
disease is likely to increase.
[00210] Optimal cellular bioenergetics rely on: (1) adequate delivery of
oxygen and substrates to
the mitochondria; (2) the oxidative capacity of mitochondria; (3) adequate
amounts of high-energy
phosphate and the creatine phosphate/ATP ratio; (4) efficient energy transfer
from mitochondria to sites
of energy utilization; (5) adequate local regulation of ATP/ADP ratios near
ATPases; and (6) efficient
feedback signaling from utilization sites to maintain energetic homeostasis in
the cell. Defects in these
cardiac energetic pathways have
67
Date Recue/Date Received 2022-04-14

been found in cardiovascular diseases such as dilated and hypertrophic
cardiomyopathies of various
origins, cardiac conduction defects, and ischemic heart diseases (Saks et al.,
J Physiol 2006, 571.2, 253-
273; Ventura-Clapier et al., J Physiol 2003, 555.1, 1-13; and Ingwall and
Weiss, Circ Res 2004, 95, 135-
145). A decrease in the creatine phosphate/ATP ratio is consistently reported
in failing human heart and
experimental heart failure, even at moderate workloads. Creatine, creatine
transporter, creatine
phosphate, and ATP are significantly reduced and the decrease in the creatine
phosphate/ATP ratio is a
predictor of mortality in congenital heart failures. Also, a down-regulation
of creatine transporter protein
expression has been shown in experimental animal models of heart disease, as
well as in failing human
myocardium, indicating that the generally lowered creatine phosphate and
creatine levels measured in
failing hearts are related to down-regulated creatine transporter capacity.
[00211] Cardiovascular disease includes hypertension, heart failure such
as congestive heart
failure or heart failure following myocardial infarction, arrhythmia,
diastolic dysfunction such as left
ventricular diastolic dysfunction, diastolic heart failure or impaired
diastolic filling, systolic dysfunction,
ischemia such as myocardial ischemia, cardiomyopathy such as hypertrophic
cardiomyopathy and dilated
cardiomyopathy, sudden cardiac death, myocardial fibrosis, vascular fibrosis,
impaired arterial
compliance, myocardial necrotic lesions, vascular damage in the heart,
vascular inflammation in the
heart, myocardial infarction including both acute post-myocardial infarction
and chronic post-myocardial
infarction conditions, coronary angioplasty, left ventricular hypertrophy,
decreased ejection fraction,
coronary thrombosis, cardiac lesions, vascular wall hypertrophy in the heart,
endothelial thickening,
myocarditis, and coronary artery disease such as fibrinoid necrosis or
coronary arteries. Ventricular
hypertrophy due to systemic hypertension in association with coronary ischemic
heart disease is
recognized as a major risk factor for sudden death, post infarction heart
failure, and cardiac rupture.
Patients with severe left ventricular hypertrophy are particularly susceptible
to hypoxia or ischemia.
[00212] Neuroprotective effects of compounds of the invention can be
determined using animal
models of cerebral ischemia such as those described, for example, in Cimino et
al., Neurotoxicol 2005,
26(5), 9929-33; Konstas et al., Neurocrit Care 2006, 4(2), 168-78; Wasterlain
et al., Neurology 1993,
43(11), 2303-10; and Zhu et al., J Neuroscience 2004, 24(26), 5909-5912.
68
Date Recue/Date Received 2022-04-14

Is chemic Reperfusion Injury
[00213] Reperfusion injury is damage to tissue when blood supply returns to
the tissue after a
period of ischemia. The absence in a tissue or organ of oxygen and nutrients
from blood creates a
condition in which the restoration of circulation results in inflammation and
oxidative damage from the
oxygen, rather than restoration of normal function. The damage of ischemic
reperfusion injury is due in
part to the inflammatory response of damaged tissue. Reperfusion contributes
to the ischemic cascade in
the brain, which is involved in stroke and brain trauma. Repeated bouts of
ischemia and reperfusion also
are believed to be a factor leading to the formation and failure to heal of
chronic wounds such as pressure
sores and diabetic foot ulcers (Mustoe, Am J Surgery 2004, 187(5), S65-S70).
In certain embodiments,
the methods and compositions of the disclosure can protect the muscle and
organs such as, for example,
the heart, liver, kidney, brain, lung, spleen and steroidogenic organs, e.g.
thyroid, adrenal glands, and
gonads, from damage as a result of ischemia reperfusion injury.
[00214] Ischemia followed by reperfusion is a major cause of skeletal and
cardiac muscle damage
in mammals. Ischemia is caused by a reduction in oxygen supplied to tissues or
organs as a result of
reduced blood flow and can lead to organ dysfunction. Reduced blood supply can
result from occlusion
or blood diversion due to vessel thrombosis, such as myocardial infarction,
stenosis, accidental vessel
injury or surgical procedures. Subsequent reestablishment of an adequate
supply of oxygenated blood to
the tissue or organ can result in increased damage, a process known as
ischemia reperfusion injury or
occlusion reperfusion injury. Complications arising from ischemia reperfusion
injury include stroke, fatal
or non-fatal myocardial infarction, myocardial remodeling, aneurysms,
peripheral vascular disease,
tissue necrosis, kidney failure, and post-surgical loss of muscle tone.
[00215] Restoration of coronary blood flow following a transient period of
ischemia (reperfusion),
though necessary for myocyte survival and to restore aerobic metabolism,
introduces a separate series of
stresses that can exacerbate cell injury. Reactive oxygen species generated
during reperfusion damage
proteins and membrane structures within cardiomyocytes and can activate signal
transduction pathways
that lead to apoptosis. Adherence of leukocytes to postischemic endothelial
cells can clog capillaries and
release inflammatory mediators. Upon reperfusion, the influx of activated
complement, catecholamines,
and other signaling molecules contained in plasma or elaborated locally within
the myocardial wall may
also influence the course of events within cells of the myocardium. As with
the direct consequences of
ischemia, reperfusion injury is an important
69
Date Recue/Date Received 2022-04-14

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
feature of acute coronary syndromes. Such injury occurs both spontaneously, as
a result of
fibrinolysis of coronary thromboses, and as a consequence of fibrinolytic
drugs of acute
angioplasty, treatments that are now commonly used to open occluded vessels.
[00216] In certain embodiments, compounds of the invention and compositions
thereof
can be used to treat a condition associated with ischemic reperfusion injury
or reduce
ischemic reperfusion injury. Ischemic reperfusion injury can be associated
with oxygen
deprivation, neutrophil activation, and/or myeloperoxidase production.
Ischemic reperfusion
injury can be the result of a number of disease states or can be
iatrogenically induced, for
example, by blood clots, stenosis or surgery.
[00217] In certain embodiments, compounds of the invention and compositions
thereof
can be used to treat stroke, a fatal or non-fatal myocardial infarction,
peripheral vascular
disease, tissue necrosis, and kidney failure, and post-surgical loss of muscle
tone resulting
from ischemic reperfusion injury. In certain embodiments, the methods and
compositions of
the invention reduce or mitigate the extent of ischemic reperfusion injury.
[00218] In certain embodiments, compounds of the invention and compositions
thereof
can be used to treat, reduce or prevent ischemic reperfusion injury associated
with occlusion
or blood diversion due to vessel stenosis, thrombosis, accidental vessel
injury or surgical
procedures.
[00219] In certain embodiments, compounds of the invention and compositions
thereof
can also be used to treat any other condition associated with ischemic
reperfusion such as
myocardial infarction, stroke, intermittent claudication, peripheral arterial
disease, acute
coronary syndrome, cardiovascular disease and muscle damage as a result of
occlusion of a
blood vessel.
[00220] In certain embodiments, compounds of the invention and compositions
thereof
can be used to treat reperfusion injury associated with myocardial infarction,
stenosis, at least
one blood clot, stroke, intermittent claudication, peripheral arterial
disease, acute coronary
syndrome, cardiovascular disease or muscle damage as a result of occlusion of
a blood vessel
[00221] In certain embodiments, compounds of the invention and compositions
thereof
can be used in conjunction with cardiac surgery, for example, in or with
cardioplegic
solutions to prevent or minimize ischemia or reperfusion injury to the
myocardium. In certain
embodiments, the methods and compositions can be used with a cardiopulmonary
bypass
machine during cardiac surgery to prevent or reduce ischemic reperfusion
injury to the
myocardium.

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
[00222] In certain embodiments, the methods and compositions of the
invention can
protect muscle and organs such as, for example, the heart, liver, kidney,
brain, lung, spleen
and steroidogenic organs, e.g. thyroid, adrenal glands, and gonads, from
damage as a result of
ischemia reperfusion injury.
[00223] Compounds and pharmaceutical compositions of the invention can be
used to
treat ischemic reperfusion injury in a tissue or organ by contacting the
tissue or organ with an
effective amount of the compound or pharmaceutical composition. The tissue or
organ may
be in a patient or outside of a patient, i.e., extracorporeal. The tissue or
organ can be a
transplant tissue or organ, and the compound or pharmaceutical composition can
be contacted
with the transplant tissue or organ before removal, during transit, during
transplantation,
and/or after the tissue or organ is transplanted in the recipient.
[00224] In certain embodiments, compounds or pharmaceutical compositions of
the
invention can be used to treat ischemic perfusion injury caused by surgery,
such as cardiac
surgery. A compound or pharmaceutical composition can be administered before,
during,
and/or after surgery. In certain embodiments, a compound or pharmaceutical
composition of
the invention can be used to treat ischemic reperfusion injury to muscle,
including cardiac
muscle, skeletal muscle or smooth muscle, and in certain embodiments, to treat
ischemic
reperfusion injury to an organ such as the heart, lung, kidney, spleen, liver,
neuron or brain. A
compound of the invention or pharmaceutical composition thereof can be
administered
before, during, and/or after surgery.
[00225] In certain embodiments, compounds of the invention or
pharmaceutical
compositions of the invention can be used to treat ischemic perfusion injury
to a muscle,
including cardiac muscle, skeletal muscle, and smooth muscle.
[00226] The efficacy of a compound of the invention for treating ischemic
reperfusion
injury may be assessed using animal models and in clinical trials. Examples of
useful
methods for assessing efficacy in treating ischemic reperfusion injury are
disclosed, for
example, in Prass et al., J Cereb Blood Flow Metab 2007, 27(3), 452-459; Arya
et al., Life
Sci 2006, 79(1), 38-44; Lee et al., Eur. J. Pharmacol 2005, 523(1-3), 101-108;
and U.S
Application No. 2004/0038891. Useful methods for evaluating transplant
perfusion/reperfusion are described, for example, in Ross et al., Am J.
Physiol-Lung Cellular
Mol. Physiol. 2000, 279(3), L528-536.
71

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
Transplant Perfusion
[00227] In certain embodiments, compounds of the invention or
pharmaceutical
compositions thereof can be used to increase the viability of organ
transplants by perfusing
the organs with a compound of the invention or pharmaceutical compositions
thereof.
Increased creatine phosphate levels are expected to prevent or minimize
ischemic damage to
an organ. Perfusing with a creatine prodrug during organ removal, following
removal of a
donor organ, during implantation, and/or following organ transplantation, can
enhance the
viability of the organ, especially a metabolically active organ, such as the
heart or pancreas,
and thereby reduce rejection rates, and/or increase the time window for organ
transplants.
[00228] In certain embodiments, compounds of the invention and compositions
thereof
can be used to treat, prevent or reduce ischemia reperfusion injury in
extracorporeal tissue or
organs. Extracorporeal tissue or organs are tissue or organs not in an
individual (also termed
ex vivo), such as in transplantation. For tissue and organ transplantation,
donor tissue and
organs removed are also susceptible to reperfusion injury during removal,
while in transit,
during implantation and following transplantation into a recipient. The
methods and
compositions can be used to increase the viability of a transplantable tissue
or organ by, for
example, supplementing solutions used to maintain or preserve transplantable
tissues or
organs. For example, the methods and compositions can be used to bathe the
transplantable
tissue or organ during transport or can be placed in contact with the
transplantable tissue or
organ prior to, during or after transplantation.
Neurodegenerative Diseases
[00229] Neurodegenerative diseases featuring cell death can be categorized
as acute,
e.g., stroke, traumatic brain injury, spinal cord injury, and chronic, e.g.,
amyotrophic lateral
sclerosis, Huntington's disease, Parkinson's disease, and Alzheimer's disease.
Although these
diseases have different causes and affect different neuronal populations, they
share similar
impairment in intracellular energy metabolism. For example, the intracellular
concentration
of ATP is decreased, resulting in cystolic accumulation of Ca2+ and
stimulation of formation
of readily oxygen species. Ca2+ and reactive oxygen species, in turn, can
trigger apoptotic cell
death. For these disorders, impairment of brain creatine metabolism is also
evident as
reflected in decreased total creatine concentration, creatine phosphate
concentration, creatine
kinase activity, and/or creatine transporter content (see e.g., Wyss and
Kaddurah-Daouk,
Physiol Rev 2000, 80, 1107-1213; Tarnopolsky and Beal, Ann Neurol 2001, 49,
561-574; and
72

Butterfield and Kanski, Mech Ageing Dev 2001, 122, 945-962).
[00230] Acute and chronic neurodegenerative diseases are illnesses
associated with high
morbidity and mortality and few options are available for their treatment. A
characteristic of many
neurodegenerative diseases, which include stroke, brain trauma, spinal cord
injury, amyotrophic lateral
sclerosis, Huntington's disease, Alzheimer's disease, and Parkinson's disease,
is neuronal-cell death. Cell
death occurs by necrosis or apoptosis. Necrotic cell death in the central
nervous system follows acute
ischemia or traumatic injury to the brain or spinal cord. It occurs in areas
that are most severely affected
by abrupt biochemical collapse, which leads to the generation of free radicals
and excitotoxins.
Mitochondrial and nuclear swelling, dissolution of organelles, and
condensation of chromatin around the
nucleus are followed by the rupture of nuclear and cytoplasmic membranes and
the degradation of DNA
by random enzymatic cuts. Apoptotic cell death can be a feature of both acute
and chronic neurological
diseases. Apoptosis occurs in areas that are not severely affected by an
injury. For example, after
ischemia, there is necrotic cell death in the core of the lesion, where
hypoxia is most severe, and apoptosis
occurs in the penumbra, where collateral blood flow reduces the degree of
hypoxia. Apoptotic cell death
is also a component of the lesion that appears after brain or spinal cord
injury. In chronic
neurodegenerative diseases, apoptosis is the predominant form of cell death.
In apoptosis, a biochemical
cascade activates proteases that destroy molecules required for cell survival
and others that mediate a
program of cell death. Caspases directly and indirectly contribute to the
morphologic changes of the cell
during apoptosis (Friedlander, N Engl J Med 2003, 348(14), 1365-75). Oral
creatine supplementation
has been shown to inhibit mitochondrial cytochrome C release and downstream
caspase-3 activation, and
ATP depletion inhibition of the caspase-mediated cell death cascades in
cerebral ischemia (Zhu et al., J
Neurosci 2004, 24(26), 5909-5912) indicating that manipulation of the creatine
kinase system may be
effective in controlling apoptotic cell death in chronic neurodegenerative
diseases.
[00231] Creatine administration shows neuroprotective effects,
particularly in animal models of
Parkinson's disease, Huntington's disease, and ALS (Wyss and Schulze,
Neuroscience 2002, 112(2), 243-
260) and it is recognized that the level of oxidative stress may be a
determinant of metabolic
determination in a variety of neurodegenerative diseases. Current hypotheses
regarding the mechanisms
of creatine-mediated neuroprotection include enhanced energy storage, as well
as stabilization of the
mitochondrial permeability transition pore by
73
Date Recue/Date Received 2022-04-14

octomeric conformation of creatine kinase. It is therefore believed that
higher levels of intracellular
creatine improve the overall bioenergetic status of a cell, rendering the cell
more resistant to injury.
Parkinson's Disease
[00232] Parkinson's disease is a slowly progressive degenerative disorder
of the nervous system
characterized by tremor when muscles are at rest (resting tremor), slowness of
voluntary movements,
and increased muscle tone (rigidity). In Parkinson's disease, nerve cells in
the basal ganglia, e.g.,
substantia nigra, degenerate and thereby reduce the production of dopamine and
the number of
connections between nerve cells in the basal ganglia. As a result, the basal
ganglia is unable to smooth
muscle movement and coordinate changes in posture, leading to tremor,
incoordination, and slowed,
reduced movement (bradykinesia) (Blandini, et al., Mol. Neurobiol. 1996, 12,
73-94).
[00233] It is believed that oxidative stress may be a factor in the
metabolic deterioration seen in
Parkinson's disease tissue (Ebadi et al., Prog Neurobiol 1996, 48, 1-19;
Jenner and Olanow, Ann Neurol
1998, 44 Suppl 1, S72-S84; and Sun and Chen, J Biomed Sci 1998, 5, 401-414)
and creatine
supplementation has been shown to exhibit neuroprotective effects (Matthews et
al., Exp Neurol, 1999,
157, 142-149).
[00234] The efficacy of administering a compound of the invention for
treating Parkinson's disease
may be assessed using animal and human models of Parkinson's disease and
clinical studies. Animal and
human models of Parkinson's disease are known (see, e.g., O'Neil et al., CNS
Drug Rev. 2005, 11(1), 77-
96; Faulkner et al., Ann. Pharmacother. 2003, 37(2), 282-6; Olson et al., Am.
J. Med. 1997, 102(1), 60-
6; Van Blercom et al., Clin Neuropharmacol. 2004, 27(3), 124-8; Cho et al.,
Biochem. Biophys. Res.
Commun. 2006, 341, 6-12; Emborg, J. Neuro. Meth. 2004, 139, 121-143; Tolwani
et al., Lab Anim Sci
1999, 49(4), 363-71; Hirsch et al., J Neural Transm Suppl 2003, 65, 89-100;
Orth and Tabrizi, Mov
Disord 2003, 18(7), 729-37; Betarbet et al., Bioessays 2002, 24(4), 308-18;
and McGeer and McGeer,
Neurobiol Aging 2007, 28(5), 639-647).
Alzheimer's Disease
[00235] Alzheimer's disease is a progressive loss of mental function
characterized by degeneration
of brain tissue, including loss of nerve cells and the development of senile
plaques and neurofibrillary
tangles. In Alzheimer's disease, parts of the brain degenerate,
74
Date Recue/Date Received 2022-04-14

destroying nerve cells and reducing the responsiveness of the maintaining
neurons to neurotransmitters.
Abnormalities in brain tissue consist of senile or neuritic plaques, e.g.,
clumps of dead nerve cells
containing an abnormal, insoluble protein called amyloid, and neurofibrillary
tangles, twisted strands of
insoluble proteins in the nerve cell.
[00236] It is believed that oxidative stress may be a factor in the
metabolic deterioration seen in
Alzheimer's disease tissue with creatine kinase being one of the targets of
oxidative damage (Pratico et
al., FASEB J 1998, 12, 1777-1783; Smith et al., J Neurochem 1998, 70, 2212-
2215; and Yatin et al.,
Neurochem Res 1999, 24, 427-435) and studies have shown a correlation between
intracellular levels of
creatine phosphate and the progress of dementia (Pettegrew et al., Neurobiol
Aging 1994, 15, 117-132).
[00237] The efficacy of administering a compound of the invention for
treating Alzheimer's
disease may be assessed using animal and human models of Alzheimer's disease
and clinical studies.
Useful animal models for assessing the efficacy of compounds for treating
Alzheimer's disease are
disclosed, for example, in Van Dam and De Dyn, Nature Revs Drug Disc 2006, 5,
956-970; Simpkins et
al., Ann NY Acad Sci, 2005, 1052, 233-242; Higgins and Jacobsen, Behav
Pharmacol 2003, 14(5-6),
419-38; Janus and Westaway, Physiol Behav 2001, 73(5), 873-86; and Conn, ed.,
"Handbook of Models
in Human Aging," 2006, Elsevier Science & Technology.
Huntington's Disease
[00238] Huntington's disease is an autosomal dominant neurodegenerative
disorder in which
specific cell death occurs in the neostriatum and cortex (Martin, N Engl J Med
1999, 340, 1970-80).
Onset usually occurs during the fourth or fifth decade of life, with a mean
survival at age onset of 14 to
20 years. Huntington's disease is fatal, and there is no effective treatment.
Symptoms include a
characteristic movement disorder (Huntington's chorea), cognitive dysfunction,
and psychiatric
symptoms. The disease is caused by a mutation encoding an abnormal expansion
of CAG-encoded
polyglutamine repeats in the protein, huntingtin. A number of studies suggest
that there is a progressive
impairment of energy metabolism, possibly resulting from mitochondrial damage
caused by oxidative
stress as a consequence of free radical generation. Preclinical studies in
animal models of Huntington's
disease have documented neuroprotective effects of creatine administration.
For example,
neuroprotection by creatine
Date Recue/Date Received 2022-04-14

is associated with higher levels of creatine phosphate and creatine and
reduced lactate levels in the brain,
consistent with improved energy production (see, Ryu et al., Pharmacology &
Therapeutics 2005, 108(2),
193-207).
[00239] The efficacy of administering a compound of the invention for
treating Huntington's
disease may be assessed using animal and human models of Huntington's disease
and clinical studies.
Animal models of Huntington's disease are disclosed, for example, in Riess and
Hoersten, U.S.
Application No. 2007/0044162; Rubinsztein, Trends in Genetics, 2002, 18(4),
202-209; Matthews et al.,
J. Neuroscience 1998, 18(1), 156-63; Tadros et al., Pharmacol Biochem Behav
2005, 82(3), 574-82, and
in U.S. Pat. No. 6,706,764, and U.S. Application Nos. 2002/0161049,
2004/0106680, and 2007/0044162.
A placebo-controlled clinical trial evaluating the efficacy of creatine
supplementation to treat
Huntington's disease is disclosed in Verbessem et al., Neurology 2003, 61, 925-
230.
Amyotrophic Lateral Sclerosis
[00240] Amyotrophic lateral sclerosis (ALS) is a progressive
neurodegenerative disorder
characterized by the progressive and specific loss of motor neurons in the
brain, brain stem, and spinal
cord (Rowland and Schneider, N Engl J Med 2001, 344, 1688-1700). ALS begins
with weakness, often
in the hands and less frequently in the feet, that generally progresses up an
arm or leg. Over time,
weakness increases and spasticity develops characterized by muscle twitching
and tightening, followed
by muscle spasms and possibly tremors. The average age of onset is 55 years,
and the average life
expectancy after clinical onset is 4 years. The only recognized treatment for
ALS is riluzole, which can
extend survival by only about three months. Oral creatine has been shown to
provide neuroprotective
effects in a transgenic animal model of ALS (Klivenyi et al., Nat Med 1999, 5,
347-50).
[00241] The efficacy of administering a compound of the invention for
treating ALS may be
assessed using animal and human models of ALS and clinical studies. Natural
disease models of ALS
include mouse models (motor neuron degeneration, progressive motor neuropathy,
and wobbler) and the
hereditary canine spinal muscular atrophy canine model (Pioro and Mitsumoto,
Clin Neurosci, 19954996,
3(6), 375-85). Experimentally produced and genetically engineered animal
models of ALS can also
useful in assessing therapeutic efficacy (see e.g., Doble and Kennelu,
Amyotroph Lateral Scler Other
Motor Neuron Disord.
76
Date Recue/Date Received 2022-04-14

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
2000, 1(5), 301-12; Grieb, Folia Neuropathol. 2004, 42(4), 239-48; Price et
al., Rev Neurol
(Paris), 1997, 153(8-9), 484-95; and Klivenyi et al., Nat Med 1999, 5, 347-
50). Specifically,
the SOD1-G93A mouse model is a recognized model for ALS. Examples of clinical
trial
protocols useful in assessing treatment of ALS are described, for example, in
Mitsumoto,
Amyotroph Lateral Scler Other Motor Neuron Disord. 2001, 2 Suppl 1, S10-S14;
Meininger,
Neurodegener Dis 2005, 2, 208-14; and Ludolph and Sperfeld, Neurodegener Dis.
2005, 2(3-
4), 215-9.
Multiple Sclerosis
[00242] Multiple sclerosis (MS) is a multifaceted inflammatory autoimmune
disease of
the central nervous system caused by an autoimmune attack against the
isolating axonal
myelin sheets of the central nervous system. Demyelination leads to the
breakdown of
conduction and to severe disease with destruction of local axons and
irreversible neuronal
cell death. The symptoms of MS are highly varied with each individual patient
exhibiting a
particular pattern of motor, sensible, and sensory disturbances. MS is
typified pathologically
by multiple inflammatory foci, plaques of demyelination, gliosis, and axonal
pathology
within the brain and spinal cord, all of which contribute to the clinical
manifestations of
neurological disability (see e.g., Wingerchuk, Lab Invest 2001, 81, 263-281;
and Virley,
NeruoRx 2005, 2(4), 638-649). Although the causal events that precipitate the
disease are not
fully understood, most evidence implicates an autoimmune etiology together
with
environmental factors, as well as specific genetic predispositions. Functional
impairment,
disability, and handicap are expressed as paralysis, sensory and octintive
disturbances
spasticity, tremor, a lack of coordination, and visual impairment, which
impact on the quality
of life of the individual. The clinical course of MS can vary from individual
to individual, but
invariably the disease can be categorized in three forms: relapsing-remitting,
secondary
progressive, and primary progressive. Several studies implicate dysfunction of
creatine
phosphate metabolism with the etiology and symptoms of the disease (Minderhoud
et al.,
Arch Neurol 1992, 49(2), 161-5; He et al,, Radiology 2005, 234(1), 211-7;
Tartaglia et al.,
Arch Neurology 2004, 61(2), 201-207; Duong et al., J Neurol 2007, Apr. 20; and
Ju et al.,
Magnetic Res Imaging 2004, 22, 427-429), although creatine ingestion alone
does not appear
to be effective in improving exercise capacity in individuals with MS (Lambert
et al., Arch
Phys Med Rehab 2003, 84(8), 1206-1210).
[00243] Assessment of MS treatment efficacy in clinical trials can be
accomplished
using tools such as the Expanded Disability Status Scale (Kurtzke, Neurology
1983, 33,
77

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
1444-1452) and the MS Functional Composite (Fischer et al., Mult Scler, 1999,
5, 244-250)
as well as magnetic resonance imaging lesion load, biomarkers, and self-
reported quality of
life (see e.g., Kapoor, Cur Opinion Neurol 2006, 19, 255-259). Animal models
of MS shown
to be useful to identify and validate potential therapeutics include
experimental
autoimmune/allergic encephalomyelitis (EAE) rodent models that simulate the
clinical and
pathological manifestations of MS (Werkerle and Kurschus, Drug Discovery
Today: Disease
Models, Nervous System Disorders, 2006, 3(4), 359-367; Gijbels et al.,
Neurosci Res
Commun 2000, 26, 193-206; and Hofstetter et al., J Immunol 2002, 169, 117-
125), and
nonhuman primate EAE models (tt Hart et al., Immunol Today 2000, 21, 290-297).
Psychotic Disorders
[00244] In certain embodiments, compounds of the invention or
pharmaceutical
compositions thereof can be used to treat psychotic disorders such as, for
example,
schizophrenia, bipolar disorder, and anxiety.
Schizophrenia
[00245] Schizophrenia is a chronic, severe, and disabling brain disorder
that affects
about one percent of people worldwide, including 3.2 million Americans.
Schizophrenia
encompasses a group of neuropsychiatric disorders characterized by
dysfunctions of the
thinking process, such as delusions, hallucinations, and extensive withdrawal
of the patients
interests from other people. Schizophrenia includes the subtypes of paranoid
schizophrenia
characterized by a preoccupation with delusions or auditory hallucinations,
hebephrenic or
disorganized schizophrenia characterized by disorganized speech, disorganized
behavior, and
flat or inappropriate emotions; catatonic schizophrenia dominated by physical
symptoms such
as immobility, excessive motor activity or the assumption of bizarre postures;
undifferentiated schizophrenia characterized by a combination of symptoms
characteristic of
the other subtypes; and residual schizophrenia in which a person is not
currently suffering
from positive symptoms but manifests negative and/or cognitive symptoms of
schizophrenia
(see DSM-IV-TR classifications 295.30 (Paranoid Type), 295.10 (Disorganized
Type),
295.20 (Catatonic Type), 295.90 (Undifferentiated Type), and 295.60 (Residual
Type),
Diagnostic and Statistical Manual of Mental Disorders, 4th Edition, American
Psychiatric
Association, 297-319, 2005). Schizophrenia includes these and other closely
associated
psychotic disorders such as schizophreniform disorder, schizoaffective
disorder, delusional
disorder, brief psychotic disorder, shared psychotic disorder, psychotic
disorder due to a
78

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
general medical condition, substance-induced psychotic disorder, and
unspecified psychotic
disorders (DSM-IV-TR, 4th Edition, pp. 297-344, American Psychiatric
Association, 2005).
[00246] Schizophrenia symptoms can be classified as positive, negative or
cognitive.
Positive symptoms of schizophrenia include delusion and hallucination, which
can be
measured using, for example, the Positive and Negative Syndrome Scale (PANSS)
(Kay et
al., Schizophrenia Bulletin 1987, 13, 261-276). Negative symptoms of
schizophrenia include
affect blunting, anergia, alogia and social withdrawal, which can be measured
for example,
using (the Scales for the Assessment of Negative Symptoms (SANS) (Andreasen,
1983,
Scales for the Assessment of Negative Symptoms (SANS), Iowa City, Iowa).
Cognitive
symptoms of schizophrenia include impairment in obtaining organizing, and
using
intellectual knowledge which can be measured using the Positive and Negative
Syndrome
Scale-cognitive subscale (PANSS-cognitive subscale) (Lindenmayer et al., J
Nery Ment Di s
1994, 182, 631-638) or by assessing the ability to perform cognitive tasks
such as, for
example, using the Wisconsin Card Sorting Test (see, e.g., Green et al., Am J
Psychiatry
1992, 149, 162-67; and Koren et al., Schizophr Bull 2006, 32(2), 310-26).
[00247] A number of studies support a correlation of schizophrenia with a
dysfunction
in brain high energy phosphate metabolism (Fukuzako, World J Biol Psychiatry
2001, 2(2),
70-82; and Gangadhar et al., Prog Neuro-Psychopharmacology & Biological
Psychiatry
2006, 30, 910-913. Patients suffering from schizophrenia exhibit lower
phosphocreatine
levels in the left and right frontal regions of the brain, which are highly
correlated with
hostility-suspiciousness and anxiety-depression assessment subscales (Deicken
et al., Biol
Psychiatry 1994, 36(8), 503-510; Volz et al., Biol Psychiatry 1998, 44, 399-
404; and Volz et
al., Biol Psychiatry 2000, 47, 954-961). Creatine supplementation has
accordingly been
proposed for treating schizophrenia (see e.g., Lyoo et al., Psychiatry Res:
Neuroimaging
2003, 123, 87-100).
[00248] The efficacy of creatine prodrugs and pharmaceutical compositions
thereof for
treating schizophrenia may be determined by methods known to those skilled in
the art For
example, negative, positive, and/or cognitive symptom(s) of schizophrenia may
be measured
before and after treatment of the patient. Reduction in such symptom(s)
indicates that a
patient's condition has improved. Improvement in the symptoms of schizophrenia
may be
assessed using, for example, the Scale for Assessment of Negative Symptoms
(SANS),
Positive and Negative Symptoms Scale (PANSS) (see, e.g., Andreasen, 1983,
Scales for the
Assessment of Negative Symptoms (SANS), Iowa City, Iowa; and Kay et al.,
Schizophrenia
Bulletin 1987, 13, 261-276), and using Cognitive Deficits tests such as the
Wisconsin Card
79

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
Sorting Test (WCST) and other measures of cognitive function (see, e.g.,
Keshavan et al.,
Schizophr Res 2004, 70(2-3), 187-194; Rush, Handbook of Psychiatric Measures,
American
Psychiatric Publishing 2000; Sajatovic and Ramirez, Rating Scales in Mental
Health, 2nd ed,
Lexi-Comp, 2003, Keefe, et al., Schizophr Res. 2004, 68(2-3), 283-97; and
Keefe et al.,
Neuropsychopharmacology, 19 Apr. 2006.
[00249] The efficacy of creatine prodrugs and pharmaceutical compositions
thereof
may be evaluated using animal models of schizophrenic disorders (see e.g.,
Geyer and
Moghaddam, in "Neuropsychopharmacology," Davis et al., Ed., Chapter 50, 689-
701,
American College of Neuropsychopharmacology, 2002). For example, conditioned
avoidance
response behavior (CAR) and catalepsy tests in rats are shown to be useful in
predicting
antipsychotic activity and EPS effect liability, respectively (Wadenberg et
al.,
Neuropsychopharmacology, 2001, 25, 633-641).
Bipolar Disorder
[00250] Bipolar disorder is a psychiatric condition characterized by
periods of extreme
mood. The moods can occur on a spectrum ranging from depression (e.g.,
persistent feelings
of sadness, anxiety, guilt, anger, isolation, and/or hopelessness,
disturbances in sleep and
appetite, fatigue and loss of interest in usually enjoyed activities, problems
concentrating,
loneliness, self-loathing, apathy or indifference, depersonalization, loss of
interest in sexual
activity, shyness or social anxiety, irritability, chronic pain, lack of
motivation, and
morbid/suicidal ideation) to mania (e.g., elation, euphoria, irritation,
and/or suspiciousness).
Bipolar disorder is defined and categorized in the Diagnostic and Statistical
Manual of
Mental Disorders, 4 th Ed., Text Revision (DSM-IV-TR), American Psychiatric
Assoc., 200,
pages 382-401. Bipolar disorder includes bipolar I disorder, bipolar II
disorder, cyclothymia,
and bipolar disorder not otherwise specified.
[00251] Patients with bipolar depression are shown to have impaired brain
high energy
phosphate metabolism characterized by reduced levels of phosphocreatine and
creatine kinase
(Kato et al., J Affect Disord 1994, 31(2), 125-33; and Segal et al., Eur
Neuropsychopharmacology 2007, 17, 194-198) possibly involving mitochondrial
energy
metabolism (Stork and Renshaw, Molecular Psychiatry 2005, 10, 900-919).
[00252] Treatment of bipolar disorder can be assessed in clinical trials
using rating
scales such as the Montgomery-Asberg Depression Rating Scale, the Hamilton
Depression
Scale, the Raskin Depression Scale, Feighner criteria, and/or Clinical Global
Impression
Scale Score (Gijsman et al., Am J Psychiatry 2004, 161, 1537-1547).

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
Anxiety
[00253] Anxiety is defined and categorized in the Diagnostic and
Statistical Manual of
Mental Disorders, 4 th Ed., Text Revision (DSM-IV-TR), American Psychiatric
Assoc., 200,
pages 429-484. Anxiety disorders include panic attack, agoraphobia, panic
disorder without
agoraphobia, agoraphobia without history of panic disorder, specific phobia,
social phobia,
obsessive-compulsive disorder, posttraumatic stress disorder, acute stress
disorder,
generalized anxiety disorder, anxiety disorder due to a general medical
condition, substance-
induced anxiety disorder, and anxiety disorder not otherwise specified. Recent
work has
documented a correlation of decreased levels of creatine/phosphocreatine in
centrum
semiovale (a representative region of the cerebral white matter) with the
severity of anxiety
(Coplan et al., Neuroimaging, 2006, 147, 27-39).
[00254] Useful animal models for assessing treatment of anxiety include
fear-
potentiated startle (Brown et al., J Experimental Psychol, 1951, 41, 317-327),
elevated plus-
maze (Pellow et al., J Neurosci. Methods 1985, 14, 149-167; and Hogg,
Pharmacol Biochem
Behavior 1996, 54(1), 21-20), and fear-potentiated behavior in the elevated
plus-maze (Korte
and De Boer, Eur J Phatinacol 2003, 463, 163-175). Genetic animal models of
anxiety are
known (Toh, Eur J Pharmacol 2003, 463, 177-184) as are other animal models
sensitive to
anti-anxiety agents (Martin, Acta Psychiatr Scand Suppl 1998, 393, 74-80).
[00255] In clinical trials, efficacy can be evaluated using psychological
procedures for
inducing experimental anxiety applied to healthy volunteers and patients with
anxiety
disorders (see e.g., Graeff, et al., Brazilian J Medical Biological Res 2003,
36, 421-32) or by
selecting patients based on the Structured Clinical interview for DSM-IV Axis
I Disorders as
described by First et al., Structured Clinical Interview for DSM-IV Axis I
Disorders, Patient
Edition (SCIDIP), Version 2. Biometrics Research, New York State Psychiatric
Institute,
New York, 1995. Any of a number of scales can be used to evaluate anxiety and
the efficacy
of treatment including, for example, the Penn State Worry Questionnaire (Behar
et al., J
Behav Ther Exp Psychiatr 2003, 34, 25-43), the Hamilton Anxiety and Depression
Scales,
the Spielberger State-Trait Anxiety Inventory, and the Liebowitz Social
Anxiety Scale
(Hamilton, J Clin Psychiatry 1980, 41, 21-24; Spielberger and Vagg, J
Personality Assess
1984, 48, 95-97; and Liebowitz, J Clin Psychiatry 1993, 51, 31-35 (Suppl.)).
Genetic Diseases Affecting the Creatine Kinase System
[00256] The intracellular creatine pool is maintained by uptake of creatine
from the
diet and by endogenous creatine synthesis. Many tissues, especially the brain,
liver and
81

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
pancreas, contain the Na+-C1- dependent creatine transport (SLC6A8), which is
responsible
for active creatine transport through the plasma membrane. Creatine
biosynthesis involves the
action of two enzymes: L-arginine:glycine amidinotransferase (AGAT) and
guanidinoacetate
transferase (GAMT). AGAT catalyses the transfer of the amidino group of
arginine to glycine
to generate ornithine and guanidinoacetate. Guanidino acetate is methylated at
the amidino
group by GAMT to give creatine (see e.g., Wyss and Kaddurah-Daouk, Phys Rev
2000, 80,
1107-213).
[00257] In humans, two genetic errors in creatine biosynthesis and one in
creatine
transporter are known and involve deficiencies of AGAT, GAMT, and creatine
transporter
(Schulze, Cell Biochem, 2003, 244(1-2), 143-50; Sykut-Cegielska et al., Acta
Biochimica
Polonica 2004, 51(4), 875-882). Patients with disorders of creatine synthesis
have systemic
depletion of creatine and creatine phosphate. Patients affected with AGAT
deficiency can
show mental and motor retardation, severe delay in speech development, and
febrile seizures
(Item et al., Am J Hum Genet. 2001, 69, 1127-1133). Patients affected with
GAMT
deficiency can show developmental delay with absence of active speech, autism
with self-
injury, extra pyramidal symptoms, and epilepsy (Stromberger et al., J Inherit
Metab Dis 2003,
26, 299-308). Patients with creatine transporter deficiency exhibit
intracellular depletion of
creatine and creatine phosphate. The gene encoding the creatine transporter is
located on the
X-chromosome, and affected male patients show mild to severe mental
retardation with
affected females having a milder presentation (Salomons et al., J. Inherit
Metab Dis 2003, 26,
309-18; Rosenberg et al., Am I Hum Genet. 2004, 75, 97-105; deGrauw et al.,
Neuropediatrics 2002, 33(5), 232-238; Clark et al., Hum Genet, 2006, April).
[00258] Creatine supplementation in dosages from about 350 mg to 2 g/kg
body
weight per day have been shown effective in resolving the clinical symptoms of
AGAT or
GAMT deficiencies (see e.g., Schulze, Cell Biochem, 2003, 244(1-2), 143-50).
However,
unlike in patients with GAMT and AGAT deficiency, in patients with creatine
transporter
deficiency oral creatine supplementation does not result in an increase in
brain creatine levels
(see Stockler-Ipsiroglu et al., in Physician's Guide to the Treatment and
Follow up of
Metabolic Diseases, eds Blau et al., Springer Verlag, 2004).
Muscle Fatigue
[00259] During high-intensity exercise, ATP hydrolysis is initially
buffered by creatine
phosphate via the creatine kinase reaction (Kongas and van Beek, 2' Int. Conf.
Systems Biol
2001, Los Angeles Calif., Omnipress, Madison, Wis., 198-207; and Walsh et al.,
J Physiol
82

2001, 537.3, 971-78). During exercise, whereas creatine phosphate is available
instantaneously for ATP
regeneration, glycolysis is induced with a delay of a few seconds, and
stimulation of mitochondrial
oxidative phosphorylation is delayed even further. Because the creatine
phosphate stores in muscle are
limited, during high-intensity exercise, creatine phosphate is depleted within
about 10 seconds. It has
been proposed that muscle performance can be enhanced by increasing the muscle
stores of creatine
phosphate and thereby delay creatine phosphate depletion. Although creatine
and/or creatine phosphate
supplementation may improve muscle performance in intermittent, supramaximal
exercise, there is no
indication that supplementation enhances endurance performance. On the other
hand, intravenous
injection of creatine phosphate appears to improve exercise tolerance during
prolonged submaximal
exercise (Clark, J Athletic Train, 1997, 32, 45-51).
Muscle Strength
[00260] Dietary creatine supplementation in normal healthy individuals has
beneficial side effects
on muscle function, and as such its use by amateur and professional athletics
has increased. There is
evidence to suggest that creatine supplementation can enhance overall muscle
performance by increasing
the muscle store of creatine phosphate, which is the most important energy
source for immediate
regeneration of ATP in the first few seconds of intense exercise, by
accelerating restoration of the creatine
phosphate pool during recovery periods, and by depressing the degradation of
adenosine nucleotides and
possibly also accumulation of lactate during exercise (see e.g., Wyss and
Kaddurah-Daouk, Physiol Rev
2000, 80(3), 1107-1213).
[00261] However, in normal healthy individuals, the continuous and
prolonged use of creatine
fails to maintain elevated creatine and creatine phosphate in muscle (see
e.g., Juhn et al., Clin J Sport
Med 1998, 8, 286-297; Terjung et al., Med Sci Sports Exerc 2000, 32, 706-717;
and Vandenberghe et
al., J Appl Physiol 1997, 83, 2055-2063), possibly as a result of the down
regulation of the creatine
transporter activity and the transporter protein content (Snow and Murphy, Mol
Cell Biochem 2001,
224(1-2), 169-181). Thus, creatine prodrugs of the invention may be used to
maintain, restore, and/or
enhance muscle strength in a mammal, and in particular a human.
83
Date Recue/Date Received 2022-04-14

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
[00262] The efficacy of administering a compound of the invention for
maintaining,
restoring, and/or enhancing muscle strength may be assessed using animal and
human models
and clinical studies. Animal models that can be used for evaluation of muscle
strength are
disclosed, for example, in Wirth et al., J Applied Physiol 2003, 95, 402-412
and Timson, J.
Appl Physiol 1990, 69(6), 1935-1945. Muscle strength can be assessed in humans
using
methods disclosed, for example, in Oster, U.S. Application No. 2007/0032750,
U.S.
Application No. 2007/0012105, and/or using other methods known to those
skilled in the art.
Organ and Cell Viability
[00263] In certain embodiments, the isolation of viable brain, muscle,
pancreatic or
other cell types for research or cellular transplant can be enhanced by
perfusing cells and/or
contacting cells with an isolation or growth media containing a creatine
phosphate analog
prodrug. In certain embodiments, the viability of a tissue organ or cell can
be improved by
contacting the tissue organ or, cell with an effective amount of a compound of
the invention
or pharmaceutical composition thereof.
Diseases Related to Glucose Level Regulation
[00264] Administration of creatine phosphate reduces plasma glucose levels,
and
therefore can be useful in treating diseases related to glucose level
regulation such as
hyperglycemia, insulin dependent or independent diabetes, and related diseases
secondary to
diabetes (U.S. Application No 2005/0256134).
[00265] The efficacy of administering a compound of the invention for
treating
diseases related to glucose level regulation may be assessed using animal and
human models
and clinical studies. Compounds can be administered to animals such as rats,
rabbits or
monkeys, and plasma glucose concentrations determined at various times (see
e.g., U.S.
Application No 2003/0232793). The efficacy of compounds for treating insulin
dependent or
independent diabetes and related diseases secondary to diabetes can be
evaluated using
animal models of diabetes such as disclosed, for example, in Shafrir, "Animal
Models of
Diabetes," Ed., 2007, CRC Press; Mordes et al., "Animal Models of Diabetes,"
2001,
Harwood Academic Press; Mathe, Diabete Metab 1995, 21(2), 106-111; and Rees
and
Alcolado, Diabetic Med. 2005, 22, 359-370.
84

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
Dose
[00266] Compounds of the invention) or pharmaceutically acceptable salts or
pharmaceutically acceptable solvates of any of the foregoing can be
administered to treat
diseases or disorders associated with a dysfunction in energy metabolism.
[00267] The amount of a compound of the invention that will be effective in
the
treatment of a particular disease, disorder or condition disclosed herein will
depend on the
nature of the disease, disorder or condition, and can be determined by
standard clinical
techniques known in the art. In addition, in vitro or in vivo assays may
optionally be
employed to help identify optimal dosage ranges. The amount of a compound
administered
can depend on, among other factors, the patient being treated, the weight of
the patient, the
health of the patient, the disease being treated, the severity of the
affliction, the route of
administration, the potency of the compound, and the judgment of the
prescribing physician.
[00268] For systemic administration, a therapeutically effective dose can
be estimated
initially from in vitro assays. For example, a dose can be formulated in
animal models to
achieve a beneficial circulating composition concentration range. Initial
doses can also be
estimated from in vivo data, e.g., animal models, using techniques that are
known in the art.
Such information can be used to more accurately determine useful doses in
humans. One
having ordinary skill in the art can optimize administration to humans based
on animal data.
[00269] Creatine occurs naturally in the human body and is partly
synthesized by the
kidney, pancreas, and liver (approximately 1-2 grams per day), and partly
ingested with food
(approximately 1-5 grams per day). Cells actively take up creatine via the
creatine
transporter. Within a cell, creatine kinase phosphorylates creatine to form a
pool of creatine
phosphate that can act as a temporal and spatial energy buffer.
[00270] Creatine, creatine phosphate, and analogs thereof can be
administered in a
high dose without adverse side effects. For example, creatine monohydrate has
been
administered to athletes and body builders in amounts ranging from 2-3 gm/day,
and creatine
phosphate has been administered to patients with cardiac diseases by
intravenous injection up
to 8 gm/day, without adverse side effects. Animals fed a diet containing up to
1%
cyclocreatine also do not exhibit adverse effects (see, e.g., Griffiths and
Walker, J. Biol.
Chem. 1976, 251(7), 2049-2054; Annesley et al., J Biol Chem 1978, 253(22),
8120-25; Lillie
et al., Cancer Res 1993, 53, 3172-78; and Griffiths, J Biol Chem 1976, 251(7),
2049-54).
[00271] In certain embodiments, a therapeutically effective dose of a
compound of the
invention can comprise from about 1 mg-equivalents to about 20,000 mg-
equivalents of a
creatine phosphate analog per day, from about 100 mg-equivalents to about
12,000 mg-

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
equivalents of creatine phosphate analog per day, from about 1,000 mg-
equivalents to about
10,000 mg-equivalents of creatine phosphate analog per day, and in certain
embodiments,
from about 4,000 mg-equivalents to about 8,000 mg-equivalents of creatine
phosphate analog
per day.
[00272] A dose
can be administered in a single dosage form or in multiple dosage
fowls. When multiple dosage forms are used the amount of compound contained
within each
dosage form can be the same or different. The amount of a compound of the
invention
contained in a dose can depend on the route of administration and whether the
disease,
disorder or condition in a patient is effectively treated by acute, chronic or
a combination of
acute and chronic administration.
[00273] In
certain embodiments an administered dose is less than a toxic dose.
Toxicity of the compositions described herein can be detei __________ mined by
standard pharmaceutical
procedures in cell cultures or experimental animals, e.g., by determining the
LD50 (the dose
lethal to 50% of the population) or the LDioo (the dose lethal to 100% of the
population). The
dose ratio between toxic and therapeutic effect is the therapeutic index. In
certain
embodiments, a pharmaceutical composition can exhibit a high therapeutic
index. The data
obtained from these cell culture assays and animal studies can be used in
formulating a
dosage range that is not toxic for use in humans. A dose of a pharmaceutical
composition of
the invention can be within a range of circulating concentrations in for
example the blood,
plasma or central nervous system, that include the effective dose and that
exhibits little or no
toxicity. A dose may vary within this range depending upon the dosage form
employed and
the route of administration utilized.
[00274] During
treatment, a dose and dosing schedule can provide sufficient or steady
state levels of an effective amount of a creatine phosphate analog to treat a
disease. In certain
embodiments, an escalating dose can be administered.
Administration
[00275] A
compound of the invention, a pharmaceutically acceptable salt, solvate,
tautomer or stereoisomer thereof or a pharmaceutically acceptable solvate of
any of the
foregoing or a pharmaceutical composition of any of the foregoing can be
administered by
any appropriate route. In certain embodiments, a compound of the invention can
be
administered intermittently or continuously. Examples of suitable routes of
administration
include, but are not limited to, intradermal, intramuscular, intraperitoneal,
intravenous,
subcutaneous, intranasal, epidural oral, sublingual, intranasal,
intracerebral, intravaginal,
86

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
transdermal, rectally, inhalation or topically. Administration can be systemic
or local.
Administration can be bolus injection, continuous infusion or by absorption
through epithelial
or mucocutaneous linings, e.g. oral mucosa, rectal, and intestinal mucosa,
etc.
[00276] In
certain embodiments, it may be desirable to introduce a compound of the
invention, a pharmaceutically acceptable salt or a pharmaceutically acceptable
solvate of any
of the foregoing or a pharmaceutical composition of any of the foregoing
directly into the
central nervous system by any suitable route, including intraventricular,
intrathecal, and
epidural injection. Intraventricular injection can be facilitated by the use
of an intraventricular
catheter, for example, attached to a reservoir, such as an Ommaya reservoir.
[00277] In
certain embodiments, a compound of the invention, a pharmaceutically
acceptable salt, solvate, tautomer or stereoisomer thereof or a
pharmaceutically acceptable
solvate of any of the foregoing or a phai ___________________________
inaceutical composition of any of the foregoing can
be administered parenterally, such as by injection, including, for example,
intravenous,
intramuscular, intraarterial, intrathecal, intracapsular, intraorbital,
intracardiac, intradermal,
intraperitoneal, transtracheal, subcutaneous, sub cuti cul ar, intraarticualr,
sub cap sul ar,
subarachnoid, intraspinal, and intrasternal injection or infusion.
[00278] A
compound of the invention, a pharmaceutically acceptable salt, solvate,
tautomer or stereoisomer thereof or a pharmaceutically acceptable solvate of
any of the
foregoing or a pharmaceutical composition of any of the foregoing can be
administered
systemically and/or locally to a specific organ.
[00279] In
certain embodiments, a compound of the invention or pharmaceutical
composition thereof can be administered as a single, one time dose or
chronically. By chronic
it is meant that the methods and compositions of the invention are practiced
more than once
to a given individual. For example, chronic administration can be multiple
doses of a
pharmaceutical composition administered to an animal, including an individual,
on a daily
basis, twice daily basis or more or less frequently, as will be apparent to
those of skill in the
art In another embodiment, the methods and compositions are practiced acutely.
By acute it
is meant that the methods and compositions of the invention are practiced in a
time period
close to or contemporaneous with the ischemic or occlusive event. For example,
acute
administration can be a single dose or multiple doses of a pharmaceutical
composition
administered at the onset of an ischemic or occlusive event such as acute
myocardial
infarction, upon the early manifestation of an ischemic or occlusive event
such as, for
example, a stroke or before, during or after a surgical procedure. A time
period close to or
contemporaneous with an ischemic or occlusive event will vary according to the
ischemic
87

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
event but can be, for example, within about 30 minutes of experiencing the
symptoms of a
myocardial infarction, stroke or intermittent claudication. In certain
embodiments, acute
administration is administration within about an hour of the ischemic event.
In certain
embodiments, acute administration is administration within about 2 hours,
about 6 hours,
about 10 hours, about 12 hours, about 15 hours or about 24 hours after an
ischemic event.
[00280] In certain embodiments, a compound of the invention or
pharmaceutical
composition thereof can be administered chronically. In certain embodiments,
chronic
administration can include several intravenous injections administered
periodically during a
single day. In certain embodiments, chronic administration can include one
intravenous
injection administered as a bolus or as a continuous infusion daily, about
every other day,
about every 3 to 15 days, about every 5 to 10 days, and in certain
embodiments, about every
days.
Combination Therapy
[00281] In certain embodiments, a compound of the invention, a
phaimaceutically
acceptable salt, solvate, tautomer or stereoisomer thereof or pharmaceutically
acceptable
solvate of any of the foregoing, can be used in combination therapy with at
least one other
therapeutic agent. A compound of the invention and other therapeutic agent(s)
can act
additively or, and in certain embodiments, synergistically. In some
embodiments, a
compound of the invention can be administered concurrently with the
administration of
another therapeutic agent, such as for example, a compound for treating a
disease associated
with a dysfunction in energy metabolism; treating muscle fatigue; enhancing
muscle strength
and endurance; increasing the viability of organ transplants; and improving
the viability of
isolated cells. In some embodiments, a compound of the invention, a
pharmaceutically
acceptable salt or a pharmaceutically acceptable solvate of any of the
foregoing can be
administered prior to or subsequent to administration of another therapeutic
agent, such as for
example, a compound for treating a disease associated with a dysfunction in
energy
metabolism such as ischemia, ventricular hypertrophy, a neurodegenerative
disease such as
ALS, Huntington's disease, Parkinson's disease or Alzheimer's disease, surgery
related
ischemic tissue damage, and reperfusion tissue damage, treating multiple
sclerosis (MS),
treating a psychotic disorder such as schizophrenia, bipolar disorder or
anxiety; treating
muscle fatigue; enhancing muscle strength and endurance; increasing the
viability of organ
transplants; and improving the viability of isolated cells.
88

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
[00282] Pharmaceutical compositions of the invention can include, in
addition to one
or more compounds of the invention, one or more therapeutic agents effective
for treating the
same or different disease, disorder or condition.
[00283] Methods of the invention include administration of one or more
compounds or
pharmaceutical compositions of the invention and one or more other therapeutic
agents
provided that the combined administration does not inhibit the therapeutic
efficacy of the one
or more compounds of the invention and/or does not produce adverse combination
effects.
[00284] In certain embodiments, compositions of the invention can be
administered
concurrently with the administration of another therapeutic agent, which can
be part of the
same pharmaceutical composition or dosage form as or in a different
composition or dosage
form from, that containing the compounds of the invention. In certain
embodiments,
compounds of the invention can be administered prior or subsequent to
administration of
another therapeutic agent In certain embodiments of combination therapy, the
combination
therapy comprises alternating between administering a composition of the
invention and a
composition comprising another therapeutic agent, e.g., to minimize adverse
side effects
associated with a particular drug. When a compound of the invention is
administered
concurrently with another therapeutic agent that potentially can produce
adverse side effects
including, but not limited to, toxicity, the therapeutic agent can
advantageously be
administered at a dose that falls below the threshold at which the adverse
side effect is
elicited.
[00285] In certain embodiments, compounds or pharmaceutical compositions of
the
invention include or can be administered to a patient together with, another
compound for
treating Parkinson's disease such as amantadine, benztropine, bromocriptine,
levodopa,
pergolide, pramipexole, ropinirole, selegiline, trihexyphenidyl or a
combination of any of the
foregoing.
[00286] In certain embodiments, compounds or pharmaceutical compositions of
the
invention include or can be administered to a patient together with, another
compound for
treating Alzheimer's disease such as donepezil, galantamine, memantine,
rivastigmine, tacrine
or a combination of any of the foregoing.
[00287] In certain embodiments, compounds or pharmaceutical compositions of
the
invention include or can be administered to a patient together with, another
compound for
treating ALS such as riluzole.
[00288] In certain embodiments, compounds or pharmaceutical compositions of
the
invention include or can be administered to a patient together with, another
compound for
89

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
treating ischemic stroke such as aspirin, nimodipine, clopidogrel,
pravastatin, unfractionated
heparin, eptifibatide, a 13-blocker, an angiotensin-converting enzyme (ACE)
inhibitor,
enoxaparin or a combination of any of the foregoing.
[00289] In certain embodiments, compounds or pharmaceutical compositions of
the
invention include or can be administered to a patient together with, another
compound for
treating ischemic cardiomyopathy or ischemic heart disease such as ACE
inhibitors such as
ramipril, captopril, and lisinopril; n-blockers such as acebutolol, atenolol,
betaxolol,
bisoprolol, carteolol, nadolol, penbutolol, propranolol, timolol, metoprolol,
carvedilol, and
aldosterone; diuretics; digitoxin or a combination of any of the foregoing.
[00290] In certain embodiments, compounds or pharmaceutical compositions of
the
invention include or can be administered to a patient together with, another
compound for
treating a cardiovascular disease such as, blood-thinners, cholesterol
lowering agents, anti-
platelet agents, vasodilators, fl-blockers, angiotensin blockers, digitalis
and is derivatives or
combinations of any of the foregoing.
[00291] In certain embodiments, compounds or pharmaceutical compositions of
the
invention include or can be administered to a patient together with, another
compound for
treating MS. Examples of drugs useful for treating MS include corticosteroids
such as
methylprednisolone; IFN-0 such as IFN-131a and IFN-131b; glatiramer acetate
(Copaxone );
monoclonal antibodies that bind to the very late antigen-4 (VLA-4) integrin
(Tysabrig) such
as natalizumab; immunomodulatory agents such as FTY 720 sphinogosie-1
phosphate
modulator and COX-2 inhibitors such as BW755c, piroxicam, and phenidone; and
neuroprotective treatments including inhibitors of glutamate excitotoxicity
and iNOS, free-
radical scavengers, and cationic channel blockers; memantine; AMPA antagonists
such as
topiramate; and glycine-site NMDA antagonists (Virley, NeruoRx 2005, 2(4), 638-
649, and
references therein; and U.S. Application No. 2004/0102525).
[00292] In certain embodiments, compounds or pharmaceutical compositions of
the
invention include or can be administered to a patient together with, another
compound for
treating schizophrenia. Examples of antipsychotic agents useful in treating
schizophrenia
include, but are not limited to, acetophenazine, alseroxylon, amitriptyline,
aripiprazole,
astemizole, benzquinamide, carphenazine, chlounezanone, chlorpromazine,
chlorprothixene,
clozapine, desipramine, droperidol, aloperidol, fluphenazine, flupenthixol,
glycine, oxapine,
mesoridazine, molindone, olanzapine, ondansetron, perphenazine, pimozide,
prochlorperazine, procyclidine, promazine, propiomazine, quetiapine,
remoxipride, reserpine,
risperidone, sertindole, sulpiride, terfenadine, thiethylperzaine,
thioridazine, thiothixene,

trifluoperazine, triflupromazine, trimeprazine, and ziprasidone. Other
antipsychotic agents useful for
treating symptoms of schizophrenia include amisulpride, balaperidone,
blonanserin, butaperazine,
carphenazine, eplavanserin, iloperidone, lamictal, onsanetant, paliperidone,
perospirone, piperacetazine,
raclopride, remoxipride, sarizotan, sonepiprazole, sulpiride, ziprasidone, and
zotepine; serotonin and
dopamine (5HT/D2) agonists such as asenapine and bifeprunox; neurokinin 3
antagonists such as
talnetant and osanetant; AMPAkines such as CX-516, galantamine, memantine,
modafinil, ocaperidone,
and tolcapone; and a-amino acids such as D-serine, D-alanine, D-cycloserine,
and N-methylglycine.
[00293] In certain embodiments, compounds or pharmaceutical compositions
of the invention
include or can be administered to a patient together with, another compound
for treating bipolar disorder
such as aripiprazole, carbamazepine, clonazepam, clonidine, lamotrigine,
quetiapine, verapamil, and
ziprasidone.
[00294] In certain embodiments, compounds or pharmaceutical compositions
of the invention
include or can be administered to a patient together with, another compound
for treating anxiety such as
alprazolam, atenolol, busipirone, chlordiazepoxide, clonidine, clorazepate,
diazepam, doxepin,
escitalopram, halazepam, hydroxyzine, lorazepam, prochlorperazine, nadolol,
oxazepam, paroxetine,
prochlorperazine, trifluoperazine, and venlafaxine.
EXAMPLES
[00295] The following examples describe in detail assays for the
characterization of compounds
of the invention and uses of compounds of the invention. It will be apparent
to those skilled in the art
that many modifications, both to materials and methods, may be practiced
without departing from the
scope of the disclosure.
General Experimental
[00296] The NMR spectra of compounds were acquired at 400 or 500 MHz (1H)
at 25 C. 1H
NMR spectra were processed with 0.3 Hz line broadening unless otherwise
specified. For LC/MS
analysis, a Shimadzu LCMS 2010 (Column: sepax ODS 50x2.0 mm, 5um), an Agilent
Tm 1200 HPLC,
1956 MSD (Column: Waters XBridge C18 4.6 x 50mm, 3.5mm) Shim-pack XR-ODS
30x3.0, 2.2um)
operating in ES (+) ionization mode, Agilent 3110TM (or an Agilent Zorbax
Bonus RP TM, 2.1 x 50mm,
3.5 um, was used. An exemplary set up was at a
91
Date Recue/Date Received 2022-04-14

temperature of 50 C and at a flow rate of 0.8 mL/min, 2 [IL injection, mobile
phase: A = water with
0.1% formic acid and 1% acetonitrile, mobile phase B = methanol with 0.1%
formic acid; retention time
given in minutes. Method details: (I) runs on a Binary Pump G1312BTM with
UVNis diode array
detector G1315C and Agilent 6130TM mass spectrometer in positive and negative
ion electrospray mode
with UV-detection at 220 and 254 nm with a gradient of 5-95% B in a 2.5 min
linear gradient (II) hold
for 0.5 min at 95% B (III) decrease from 95-5% B in a 0.1 min linear gradient
(IV) hold for 0.29 min at
5% B. For analytical HPLC sample analysis, an Agilent 1200 SeriesTM was used
equipped with a Waters
HSS T3Tm column, 2.1 x 50mm, 1.8 jtm, at a temperature of 60 C and at a flow
rate of 0.5 mL/min,
mobile phase: A = water with 0.1% formic acid and 0.1% acetonitrile, mobile
phase B = acetonitrile with
0.1% formic acid; retention time given in minutes. Melting point temperatures
were recorded using a
Thomas Hoover UnimeltTM capillary melting point apparatus. Reaction progress
was monitored by thin
layer chromatography on Merck'' EMD 60 F254 silica gel coated glass plates
using UV light and/or
treatment with iodine to visualize. Chromatographic purification was carried
out on either a Teledyne'
ISCO CombiFlash CompanionTM with a variable flow rate from 5-100 mL/min. The
columns used were
Teledyne ISCO RediSep Disposable Flash Columns (4, 12, 24, 40, 80, or 120 g
pre-packed silica gel).
Peaks were detected by variable wavelength UV absorption (200-360 nm).
Preparative reverse phase
chromatography was accomplished using a Gilson 215 Liquid Handler equipped
with Varian Model 218
pumps operated using ChromeleonTM software. Detection was achieved using
either a Varian Pro StarTM
UV-Vis or a Sedex 55TM ELSD unit. Chromatographic separations were achieved
using a Phenomenex
KinetexTM 5u C18 100A, Axia, 100x30 mm column at a flow rate of 28 mLmin-1.
[00297] The compounds tested in the bioassays, such as Compounds A, B, C,
D, E, F, G, H, J, K,
L, and M, correspond to the compounds exemplified by the synthetic procedures
described herein. For
example, Compound E is the compound of Example 26, Step 5A as described in
this application.
Example 1: Methods for Determination of Enzymatic Cleavage of Prodrugs In
Vitro
[00298] For creatine prodrugs, it is generally desirable that the prodrug
remains intact (i.e.,
uncleaved) while in the systemic circulation and be cleaved (i.e., to release
the parent drug) in the target
tissue. A useful level of stability can at least in part be determined by the
mechanism and
pharmacokinetics of the prodrug. A useful level of lability can at least in
part also be determined by the
pharmacokinetics of the prodrug and parent drug (e.g., creatine) in
92
Date Recue/Date Received 2022-04-14

the systemic circulation and/or in the gastrointestinal tract, if orally
administered. In general, prodrugs
that are more stable in the gastrointestinal tract (as may be assessed by
stability in simulated gastric fluid,
simulated intestinal fluid, intestinal S9, pancreatin or colonic wash assays)
and are more labile in mouse
plasma, rat plasma, human plasma, mouse, rat and/or human liver S9, liver
microsomes, and/or
hepatocyte preparations can be useful as an orally administered prodrug. In
general, prodrugs that are
more stable in mouse plasma, rat plasma, human plasma, mouse, rat and/or human
liver S9, liver
microsomes, and/or hepatocyte preparations and which are more labile in target
tissue cell homogenates
or target tissue cell isolate preparations, such as brain, muscle, and Caco-2
S9 preparations, can be useful
as systemically administered prodrugs and/or can be more effective in
delivering a prodrug to a target
tissue. In general, prodrugs that are more stable in different pH
physiological buffers can be more useful
as prodrugs. In general, prodrugs that are more labile in target tissue cell
homogenates and/or target tissue
cell isolate preparations, such brain, muscle and Caco-2 S9 preparations, can
be intra-cellularly cleaved
to release the parent drug to a target tissue. The results of tests, such as
those described in this example,
for determining the enzymatic or chemical cleavage of prodrugs in vitro can be
used to select prodrugs
for in vivo testing.
[00299] The stabilities of prodrugs can be evaluated in one or more in
vitro systems using a variety
of preparations following methods known in the art. Tissues and preparations
are obtained from
commercial sources (e.g., Pel-Freez Biologicals', Rogers, Ark., or GenTest
Corporation, Woburn,
Mass.). Experimental conditions useful for the in vitro studies are described
in Table 1. Prodrug is added
to each preparation in triplicate.
Table 1: Standard' Conditions for Prodrug In Vitro Stability and Metabolism
Studies
Enzyme/Protein Substrate
Assay Cofactors
Concentration Concentration (I'M)
SGF +/- 0.1 mg/mL pepsin 1 - 10 NA
+1- 1 % w/v
SIF 1 - 10 NA
pancreatin
Plasma NA 1 - 10 +/- DIFP, 10 mM'
Blood NA 1 - 10 +/- DIFP, 10 mM'
Liver microsomes 0.5 mg/mL 1 - 10 +/- NADPHb
Liver or Intestinal S9 1 mg/mL 1 - 10 +/- NADPHb
Hepatocytes NA 1 ¨ 10 NA
Tissue homogenate' NA 1 ¨ 10 NA
'Typical test range provided, range may be exceeded dependent upon intended
clinical use of prodrug;
bNADPH generating system, e.g., 1.3 mM NADP+, 3.3 mM glucose-6-phosphate, 0.4
U/mL glucose-6-
phosphate dehydrogenase, 3.3 mM magnesium chloride and 0.95 mg/mL potassium
phosphate, pH 7.4;
'Examples: brain tissue homogenate, muscle tissue homogenate; dAssay can be
performed with and
without addition of diisopropyl fluorophosphonate (DIFP, serine protease
inhibitor) to determine if
degradation is mediated by a serine protease, e.g., carboxylesterase (CES)
93
Date Recue/Date Received 2022-04-14

[00300] For preparations that contain alkaline phosphatases, prodrug is
tested in the presence and
absence of a phosphatase inhibitor cocktail (Sigma'). Samples are incubated at
37 C for times ranging
from 30 minutes to 24 hours. At each time point, samples are quenched with 50%
ethanol. Baseline
concentrations of prodrug are determined by adding the compound directly to
the 50%
ethanol/preparation mixture (t=0). Samples are centrifuged at 14,000 rpm for
15 minutes, and
concentrations of intact prodrug and released parent drug are determined using
LC/MS/MS. This stability
of prodrugs towards specific enzymes (e.g., peptidases, etc.) is also assessed
in vitro by incubation with
the purified enzyme.
[00301] Pancreatin stability studies are conducted by incubating prodrug (5
uM) with 1% (w/v)
pancreatin (Sigma, P-1625, from porcine pancreas) in 0.025 M Tris buffer
containing 0.5 M NaCl (pH
7.5) at 37 C. The reaction is stopped by addition of 3 volumes of 50%
ethanol. After centrifugation at
14,000 rpm for 15 min, the supernatant is removed and analyzed by LC/MS/MS for
prodrug, creatine,
and creatinine.
[00302] For determination of stability in simulated gastric fluid (SGF),
prodrug (10 M) is
incubated in SGF (0.2% NaCl w/v, 0.7 % HC1 v/v, pH1.2) with and without
addition of pepsin (3.2 g of
purified pepsin per liter with an activity of 800 - 2500 units per mg of
protein) at 37 C. At selected time
points (e.g., 0, 15, 30, 60 and 120 min) 50 L aliquots are removed and
neutralized by addition of 50 L
of 0.1M sodium bicarbonate solution, followed by 150 1.1L of ice-cold
acetonitrile. Samples are
centrifuged at 4,000 x g for 15 min at 4 C, and the supernatants are removed
and analyzed for prodrug,
creatine and creatinine concentrations by LC-MSMS (Table 2).
Table 2: Stability of Prodrugs Simulated Gastric Fluid (SGF) with and without
Pepsin
T112 (min)
Compound of
Compound ID
Formula (+) pepsin (-) pepsin
A III 117 >360
B III 133 >360
C III >360 >360
D III >360 >360
E III >360 >360
F III >360 >360
G III >360 >360
[00303] For determination of stability in simulated intestinal fluid (SIF),
prodrug (10 uM) is
incubated in SIF (0.68 % KH2PO4 w/v, 0.86 % NaOH v/v, pH 6.8) with and without
94
Date Recue/Date Received 2022-04-14

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
addition of pancreatin (1 % w/v) at 37 C. At selected time points (e.g., 0,
15, 30, 60 and 120
min) 50 uL aliquots are removed and the reactions are terminated by addition
of 150 uL of
ice-cold acetonitrile. Samples are centrifuged at 4,000 x g for 15 min at 4
C, and the
supernatants are removed and analyzed for prodrug, creatine and creatinine
concentrations by
LC-MSMS (Table 3).
Table 3: Stability of Prodrugs Simulated Intestinal Fluid (SIF) with and
without Pancreatin
Compound of T1/2 (min)
Compound ID
Formula
(+) pepsin (-) pepsin
A III >360 >360
B III >360 >360
C III >360 >360
D III >360 >360
E III >360 >360
F III >360 >360
G III >360 >360
[00304] To determine stability in Caco-2 homogenate S9, Caco-2 cells are
grown for
21 days prior to harvesting. Culture medium is removed and cell monolayers are
rinsed and
scraped off into ice-cold 10 mM sodium phosphate/0.15 M potassium chloride, pH
7.4. Cells
are lysed by sonication at 4 C using a probe sonicator. Lysed cells are then
transferred into
1.5 mL centrifuge vials and centrifuged at 9,000 g for 20 min at 4 C. The
resulting
supernatant (Caco-2 cell homogenate S9 fraction) is aliquoted into 0.5 mL
vials and stored at
-80 C until used.
[00305] For stability studies, prodrug (5 NI) is incubated in Caco-2
homogenate S9
fraction (0.5 mg/mL in 0.1M Tris buffer, pH 7.4) at 37 C. Triplicate samples
are quenched at
each time point with 50% ethanol. The initial (t=0) concentration of prodrug
is determined by
adding 5 jiM prodrug directly to a 50% ethanol/Caco-2 homogenate mixture.
Samples are
subjected to LC/MS/MS analysis to determine concentrations of prodrug,
creatine and
creatinine.
[00306] To determine prodrug stability in mouse, rat, human or plasma from
other
species, prodrugs (10 uM) or positive controls (10 uM, propantheline or
procaine) are
incubated in undiluted plasma. Duplicate samples of prodrugs and controls are
prepared and
analyzed. Stock solutions of prodrugs are prepared in DMSO (10 mM) and diluted
to 0.1

mM in pH 7.4 phosphate buffer to prepare spiking solutions. The prodrug
spiking solutions are aliquoted
(10 L) into 96-well plates. Pre-warmed (37 C) plasma (90 L) is added to the
wells designated for 5,
15, 30,45 and 60 min time points; fort = 0 min the quench solution (400 L
acetonitrile) is added directly
to the prodrug containing well followed by 90 L of pre-warmed plasma. At 5,
15, 30, 45 and 60 min
400 L aliquots of acetonitrile are added to the wells to stop the reaction.
After quenching, the plates
are shaken for 10 min (600 rpm) and then centrifuged at 5500 g for 15 min.
Aliquots (50 L) are
transferred to the analysis plate and diluted with100 L ultrapure water
(Millipore) for LC-MSMS
quantitation of prodrug concentrations, and in some cases creatine and/or
creatinine. Chromatography
columns (e.g., Atlantis' HILIC Silica, Gemini C-18, Ultimate XB-C18) are
selected based upon the
lipophilicity and polarity of each prodrug. LC-MSMS platforms (e.g., SciexTm
API4000, Sciex
API6500) are also selected based on the requirements of each prodrug. The
stabilities of the compounds
of the present disclosure (prodrugs) in mouse plasma incubations is shown in
Table 4 and stability in
human plasma incubations is shown in Table 5.
Table 4: Stability of Selected Compounds in Mouse Plasma Incubations
Compound Compound of % Remaining
T1/2 (min)
ID Formula
min 60 min
A jjj 102 94.0 > 180
B III 110 96.0 >180
85.0 114 >180
124 118 >180
E III 100 89.0 > 180
F III 103 88.2 118
G III 101 91.1 >180
H III 96.6 94.5 > 180
VI 53.8 <LOD 3.32
VI 37.2 0.08 NC
97.9 101 > 180
LOD = Limit of detection
Concentration of compounds (prodrugs) and positive control: 10 M
Duplicative samples per time point obtained
96
Date Recue/Date Received 2022-04-14

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
Table 5: Stability of Selected Compounds in Human Plasma Incubations
Compound Compound of % Remaining
T1/2 (min)
ID Formula
min 60 min
A jjj 105 88.8 >180
B III 97.4 87.8 >180
86.5 119 131
100 109 138
In 101 75.6 154
F III 92.7 68.1 > 180
In 115 90.8 > 180
H III 114 109 >180
VI 56.4 0.01 4.32
VI 42.5 0.05 3.16
96.1 91.6 >180
Concentration of compounds (prodrugs) and positive control: 10 uM
Duplicative samples per time point obtained
[00307] For liver microsomal stability studies, prodrug or positive control
(testosterone, propafenone, diclofenac, 7-ethoxycoumarin or propranol ol) are
incubated (in
duplicate) at 5 tiM in hepatic or intestinal fractions from mouse, human, dog,
monkey and/or
rat. Incubations are conducted at 37 C in the presence or absence of NADPH
regenerating
system to indicate whether metabolism proceeds via an NADPH requiring enzyme
(i.e.
P450s, FM0s, NADPH-P450 reductase or other oxidase enzymes). Duplicate samples
of
prodrugs and controls are prepared and analyzed. Stock solutions of prodrugs
are prepared in
DMSO (10 mM) and diluted to 0.05 mM in a mixture of 25 % Me0H/pH 7.4 phosphate
buffer to prepare spiking solutions. The prodrug spiking solutions are
aliquoted (10 !IL) into
96-well plates. Pre-warmed (37 C) microsome solution (80 L) is added to the
wells
designated for 5, 15, 30, 45 and 60 min time points and incubated for 10 min
before initiating
the reaction with 10 jiL of NADPH regenerating solution. For t = 0 min the
quench solution
(300 ILIL acetonitrile) is added directly to the prodrug containing well
followed by the
microsome solution and NADPH soluton. Incubations of the prodrugs in heat-
inactivated
fractions or buffer are conducted to differentiate enzymatic from non-
enzymatic degradation.
At specified time points (e.g., 0, 5, 10, 20, 30 and 60 min), samples are
taken and teiminated
with an equal volume of cold acetonitrile containing a suitable internal
standard (e.g.,
97

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
labetalol, tolbutamide). After quenching, the plates are centrifuged at 4000 g
for 20 min.
Aliquots (100 ttL) are transferred to the analysis plate and diluted with 400
ttL ultrapure
water (Millipore) for LC-MSMS quantitation of prodrug concentrations, and in
some cases
creatine and/or creatinine. Chromatography columns (e.g., ACE 5 Phenyl,
Phenomenex C18
Synergi Hydro-RP, Atlantis HILIC Silica) are selected based upon the
lipophilicity and
polarity of each prodrug. LC-MSMS platforms (e.g., Sciex API4000, Sciex
API6500) are
also selected based on the requirements of each prodrug. The metabolic
stability of the
compounds of the present disclosure of in mouse liver microsomal incubations
is shown in
Table 6 and human liver microsomal incubations is shown in Table 7.
Table 6: Metabolic Stability of Selected Compounds in Mouse Liver Microsomal
Incubations
Compound Compound % Remaining % Remaining
of (w/ NADPH) (No
NADPH) T (min) in vivo Clint
ID 1/2
(ml/min/kg)
Formula 5 min 30 min 30 min
A III 109 112 111 >90 <71
B III 109 103 99.0 >90 <71
C III 109 112 103 >90 <71
D III 89.3 69.5 91 34.7 183
E III 89.1 26.0 15.6 20.0 407
F III 111 94.7 309 >90 <71
G III 124 96.9 223 >90 <71
H III 126 107 125 >145 <38
J VI 25.3 0.10 1.3 4.7 1160
L I 76.6 53.5 72.3 33.2 165
M I NA 41.5 52.6 19.6 280
Concentration of compounds (prodrugs) and positive control: 10 ttM
Duplicative samples per time point obtained
98

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
Table 7: Metabolic Stability of Selected Compounds in Human Liver Microsomal
Incubations
Compound Compound A Remaining A Remaining
of (w/ NADPH) (No
NADPH) T (mm) in vivo Clint
ID 1/2 (ml/min/kg)
Formula 5 min 30 min 30 min
A
114.2 99.2 89.0 >90 <21
III
B III 96.8 85.5 91 >90 <21
C
112.8 76.2 103 74.4 25
III
D
83.6 91.9 108 >90 51
III
E
103.8 82.1 106 >90 <21
III
F III 102.8 90.8 85.0 >90 <21
G
111.2 83.0 103 >90 <21
III
H III 66.5 64.9 72.4 130 9.60
J VI 38.9 1.80 5.0 11.2 111
L I 64.5 48.1 61.1 20.9 59.8
M I 64.1 38.8 _ 56.6 21.5 57.9
Concentration of compounds (prodrugs) and positive control: 10 tiM
Duplicative samples per time point obtained
[00308] For S9
stability studies, prodrug (5 uM) is incubated in mouse, human, dog,
monkey and/or rat liver or intestinal S9 homogenate (0.5 mg/mL in 0.1M
potassium
phosphate buffer, pH 7.4, 1 mM NADPH) at 37 C. Incubations are conducted in
the
presence or absence of NADPH regenerating system to indicate whether
metabolism
proceeds via an NADPH requiring enzyme (i.e. P450s, FM0s, NADPH-P450 reductase
or
other oxidase enzymes). Triplicate samples are quenched at each time point
with 50%
ethanol. The initial (t=0) concentration of prodrug is determined by adding 5
uM prodrug
directly to a 50% ethanol/S9 homogenate mixture. Samples are subjected to
LC/MS/1\4S
analysis to determine concentrations of prodrug, creatine, and creatinine.
[00309] For
hepatocyte stability studies, prodrug (5 uM) is incubated with plated
hepatocytes (e.g., mouse, rat, human). Fresh hepatocytes are received (Life
Technologies)
plated in a 12-well format with overlay (except rat which has no overlay).
Upon receipt,
shipping media is removed immediately and replaced with 1 mL pre-warmed
culture
99

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
medium. Cells are acclimated overnight at 37 C with 5% CO2 atmosphere. Media
is
aspirated from the plate and replaced with 1 mL fresh media containing prodrug
(5 1.tM) or
solvent control (0.0125% DMSO). Samples (triplicate)are incubated at 37 C in
5% CO2
atmosphere for 0, 0.25, 0.5, 0.75, 1, 2 and 4 hr. Extra wells with solvent
control are included
for production of calibration curves and measurement of background. At the
selected time
point, media is removed and frozen. Cells are washed twice with cold PBS. 0.5
mL cold 70%
acetonitrile containing internal standard is added to each well and cells are
gently removed
from the plate by scraping. The recovered cells suspended in the organic
solution are
aspirated into a vial and frozen at -80 C. For analysis, cell solutions in 70%
ACN are
removed from the freezer, defrosted and vortexed. 500 [IL water is added to
each tube and
the samples are vortexed again. Tubes are centrifuged at 13000 rpm for 10
minutes at 4 C.
Cell supernatants and the original recovered media are removed and analyzed by
LC-MS/MS
for determination of prodrug, creatine, and creatinine.
[00310] Three
buffers are used to determine the chemical stability of prodrug: (1)
0.1M potassium phosphate, 0.5 M NaCl, pH 2.0, (2) 0.1 M Tris-HC1, 0.5 M NaCl,
pH 7.4,
and (3) 0.1 M Tris-HCl, 0.5 M NaCl, pH 8Ø Prodrug (5 ttM) is added to each
buffer in
triplicate. Samples are quenched at each time point with 50% ethanol. The
initial (t=0)
concentration of prodrug is determined by adding 5 1,t1V1 prodrug directly to
a 50% ethanol/pH
Buffer mixture. Samples are subjected to LC/MS/MS analysis to determine
concentrations of
prodrug, creatine and creatinine.
Example 2: In Vitro Determination of Release of Creatine from Prodrugs
[00311] For
assessment of the ability of prodrugs to release creatine, and to release
creatine preferentially to unwanted cyclization to creatinine, d3-labeled
(deuterium labeled
methyl group) prodrugs are incubated with liver homogenates (e.g., mouse,
human) specially
prepared to preserve N-reductase activity. The use of d3-labelled prodrugs is
essential in
order to distinguish prodrug-derived creatine (d3-creatine) from high
concentrations of
endogenous (non-labeled) creatine. Incubations (37 C) are performed in 100
m114 potassium
phosphate buffer, pH 6.0 to optimize N-reductase activity. Prodrugs are tested
at final
concentrations of 20 ttM and 200 [IM. Approximately 4-5 mg of homogenate is
used for
each reaction. Co-factor (NADH) is included at a final concentration of 1 mM.
Benzamidoxime (500 ttM final concentration) is used as a positive control for
N-reductase
activity. N-
reductase activity is confirmed by conversion of benzamidoxime into
benzamidine. Negative controls include incubations without NADH (to assess
NADH-
100

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
independent prodrug cleavage) and incubations without liver homogenate (but
with NAPD)
to assess non-enzymatic prodrug cleavage under the conditions of the assay.
Prodrug
incubations are prepared by addition of 10 L of prodrug stock solution (400
or 4000 M, 40
% DMSO in water) to 100 L of the potassium phosphate buffer, followed by
addition of 70
1.iL of liver homogenate. Reaction is initiated by addition of 20 1.it of NADH
solution (10
mM), or 20 !AL water for (¨)NADH negative controls. At selected time points
(e.g., 0, 30, 60
and 180 min) 50 1.it aliquots are removed and the reactions are tenninated by
addition of 150
p.L of ice-cold acetonitrile (80% ACN/20% water) stop solution. The samples
are
centrifuged at 15890 x g for 10 minutes at 4 C, followed by transfer of the
supernatants for
storage at 40 C pending LC-MSMS analysis. Sample supernatants are analyzed by
LC-
MSMS (HILIC column) for determination of d3-prodrug, d3-creatine and d3-
creatinine levels
(Table 8).
Table 8: In Vitro Release of d3-Creatine and d3-Creatinine from d3-Prodrugs in
Mouse Liver
Homogenate (MLH) Incubations
Compound Compound of Incubation Concentration (pM) at 120 min
ID Formula Conditions Prodrug d3-
Creatine d3-Creatinine
Prodrug +MLH
195 45.4 ND
+NADH
Negative Control
A III 254 15.0 ND
(No NADH)
Negative Control
361 0.29 ND
(No MLH)
Prodrug +MLH
4.70 39.1 ND
+NADH
Negative Control
E III ND 10.0 ND
(No NADH)
Negative Control
240 ND ND
(No MLH)
Prodnig +MLH
157 9.68 19.9
+NADH
Negative Control
F III 176 2.25 4.52
(No NADH)
Negative Control
259 ND ND
(No MLH)
ND = not determined
Example 3: In Vitro Determination of Caco-2 Cellular Permeability of Prodrugs
[00312] The passive
permeability of creatine prodrugs is assessed in vitro using
standard methods well known in the art (see, e.g., Stewart, et al., Pharm.
Res., 1995, 12, 693).
101

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
For example, passive permeability can be evaluated by examining the flux of a
prodrug
across a cultured polarized cell monolayer (e.g., Caco-2 cells).
[00313] Caco-2 cells obtained from continuous culture (passage less than
28) are
seeded at high density onto Transwell polycarbonate filters. Cells are
maintained with
DMEM/10% fetal calf serum+0.1 mM nonessential amino acids+2 mM L-Gln, 5%
CO2/95%
02, 37 C until the day of the experiment. Permeability studies are conducted
at pH 6.5
apically (in 50 mM MES buffer containing 1 mM CaCl2, 1 mM MgCl2, 150 mM NaCl,
3
mM KC1, 1 mM NaH2PO4, 5 mM glucose) and pH 7.4 basolaterally (in Hank's
balanced salt
solution containing 10 mM I-1EPES) in the presence of efflux pump inhibitors
(250 hM MK-
571, 250 p..M verapamil, 1 mM Ofloxacin). Inserts are placed in 12 or 24 well
plates
containing buffer and incubated for 30 min at 37 C. Prodrug (100 hM, 250 hM,
300 hM or
500 M) is added to the apical or basolateral compartment (donor) and
concentrations of
prodrug and/or released parent drug (creatine) in the opposite compartment
(receiver) are
determined at intervals over 1 hour using LC/MS/MS. Values of apparent
permeability (P app)
app
are calculated using the equation:
Papp¨VddC/cit)/(AC0)
where Vr is the volume of the receiver compartment in mL; dC/dt is the total
flux of prodrug
and parent drug ( M/s), determined from the slope of the plot of concentration
in the receiver
compartment versus time; Co is the initial concentration of prodrug in hM; and
A is the
surface area of the membrane in cm2. In certain embodiments, prodrugs with
significant
transcellular permeability exhibit a value of Papp of > 1 x10-6 cm/s, in
certain embodiments, a
value of Papp of > 1 x10-5 cm/s, and in certain embodiments a value of Papp of
>5 x10-5 cm/s.
Example 4: Uptake by Caco-2 and HEK-2 Cells
[00314] Caco-2 or HEK Peaks are seeded onto poly-lysine coated 24-well
plastic cell
culture plates at 250,000 and 500,000 cells/well, respectively. Cells are
incubated overnight
at 37 C. Prodnig is added to each well in 1 mL fresh media. Each
concentration of prodrug is
tested in triplicate. Media only is added to the control wells. At each time
point, cells are
washed four times in Hank's Balanced Salt Solution. Cells are lysed and
compound is
extracted by adding 200 [IL 50% ethanol to each well for 20 minutes at room
temperature.
Aliquots of the ethanol solution are moved to a 96-well V-bottom plate and
centrifuged at
5,700 rpm for 20 minutes at 4 C. Supernatant is analyzed by LC/MS/MS to
determine the
concentration of prodrug, creatine, and/or creatinine.
102

Example 5: Expression of SMVT in Mammalian Cells
[00315] Sodium dependent multivitamin transporter (SMVT; product of the
SLC5A6 gene)was
subcloned into a plasmid that allows for inducible expression by tetracycline
(TREX plasmid, Invitrogen
Inc., Carlsbad Calif.). The SMVT expression plasmid was transfected into a
human embryonic kidney
(HEK) cell line and stable clones were isolated by G418 selection and flow
activated cell sorting (FACS).
Biotin uptake in a SMVT-HEK cell clone was used for validation. SMVT-HEK/TREX
cells were plated
in 96-well plates at 100,000 cells/well at 37 C for 24 hours and tetracycline
(1 g/mL) was added to
each well for an additional 24 hours to induce SMVT transporter expression.
Radiolabeled 3H-biotin
(.about.100,000 cpm/well) was added to each well. Plates were incubated at
room temperature for 10
min. Excess 3H-biotin was removed and cells were washed three times with a 96-
well plate washer with
cold assay buffer. Scintillation fluid was added to each well, and the plates
were sealed and counted in a
96-well plate-based scintillation counter.
[00316] Similar methods can be used to prepare HEK cells expressing other
transporters or other
cell lines expressing SMVT or other transporters.
[00317] The GenBank accession number for human SMVT is NM_021095.
Reference to the
SMVT transporter includes the amino acid sequence described in or encoded by
the GenBank reference
number NM_021095, and, allelic, cognate and induced variants and fragments
thereof retaining
essentially the same transporter activity. Usually such variants show at least
90% sequence identity to
the exemplary GenBank nucleic acid or amino acid sequence. Substrates for SMVT
are compounds
containing a free carboxylic acid and a short alkyl chain, e.g., C1_6 alkyl,
ending in a cyclic or branched
group. Example of SMVT substrates include biotin, pantothenic acid, and 4-
phenylbutyric acid.
Example 6: Competition Assays Using SMVT
[00318] To determine if a creatine prodrug binds the SMVT transporter, a
competition binding
assay was developed. This assay measures how different concentrations of a
test compound block the
uptake of a radiolabeled substrate such as biotin or pantothenic acid. The
half-maximal inhibitory
concentration (IC50) for inhibition of transport of a substrate by a test
compound is an indication of the
affinity of the test compound for the SMVT transporter. If the test compound
binds SMVT competitively
with the radiolabeled substrate, less of the radiolabeled substrate is
transported into the HEK cells. For
test compounds that do not interact with SMVT in a manner competitive with
substrates the curve
remains an essentially
103
Date Recue/Date Received 2022-04-14

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
flat line, i.e., there is no dose response seen. The amount of radiolabeled
substrate taken up
by the cells is measured by lysing the cells and measuring the radioactive
counts per minute.
Competition binding studies are performed as follows. SMVT-HEK/TREX cells are
plated in
96-well plates at 100,000 cells/well at 37 C for 24 hours and tetracycline (1
tig/mL) is
added to each well for an additional 24 hours to induce SMVT transporter
expression.
Radiolabeled 3H-biotin (about.100,000 cpm/well) is added to each well in the
presence and
absence of various concentrations of unlabeled biotin or pantothenic acid in
duplicate or
triplicate. Plates are incubated at room temperature for 10 min. Excess 3H-
biotin is removed
and cells are washed three times using a 96-well plate washer with cold assay
buffer.
Scintillation fluid is added to each well, and the plates are sealed and
counted in a 96-well
plate-based scintillation counter. Data is graphed and analyzed using non-
linear regression
analysis with Prism Software (GraphPad, Inc., San Diego, Calif.).
Example 7: Treatment of HEK SMVT Cells with Creatine Prodrugs
[00319] Uptake of unlabeled creatine prodrugs is measured in HEK cells
stably
expressing SMVT. Cells are plated at a density of 250,000 cells/well in
polylysine coated 24-
well tissue culture plates. Twenty-four hours later cells are treated with
tetracycline (1 [tg/m1)
to induce SMVT expression or left untreated. The following day (approximately
48 hours
after seeding), the assay is performed. Creatine prodrugs (0.1 mM final
concentration) are
added to a buffered saline solution (HBSS), and 0.5 mL of each test solution
is added to each
well. Cells are allowed to take up the test compounds for 1 or 3 hours. Test
solution is
aspirated and cells washed 4 times with ice-cold HBSS. Cells are then lysed
with a 50%
ethanol solution (0.2 mL/well) at room temperature for 15 minutes. The lysate
is centrifuged
at 5477 x g for 15 minutes at 4 C to remove cell debris. The concentration of
creatine
prodrugs and creatine in the cell is determined by analytical LC/MS/MS.
Transporter specific
uptake is determined by comparison with control cells lacking transporter
expression.
Example 8: Effect of Treatment on the Creatine Kinase System
[00320] HEK cells expressing SMVT are treated with buffer, a creatine
prodrug (100
[tM), creatine (100 tiM) or creatine analog (100 !LIM), for a specified time
period according to
the protocol of Example 6. Following treatment, the intracellular
concentrations of the
creatine prodrug, creatine phosphate, ATP, and creatine and/or creatine analog
are measured
by analytical LC/MS/MS.
104

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
Example 9: Restoration of Cellular Energy Homeostasis Following Sodium Azide
Treatment
[00321] An adaptation of the methods described by Weinstock and Shoham,
Neural
Transm. 2004, 111(3), 347-66, is used to evaluate the protective effects on
intracellular
energy homeostasis of compounds of the invention.
[00322] The HEK TREX SMVT cell line is seeded at 250 k per well in a 24-
well
polylysine coated tissue culture plate. The next day, cells are treated with
doxycycline (1
lig/mL) to express the SMVT transporter, which is required for efficient
uptake of the
creatine prodrug, e.g., a compound of the invention, tested. The cells are
incubated and
assayed on the following day. Cells are washed twice with HBSS buffer lacking
glucose.
Cells are then incubated for 20 mM at 37 C in a 5% CO2 incubator in the same
buffer with or
without sodium azide. A typical range of sodium azide used in these
experiments is from 1
mM to 9 mM. After this time, 300 tiM of a prodrug of a creatine analog is
added to the cells
or the cells are left untreated. In some experiments, creatine is used as a
comparison. The
cells are incubated for an additional 20 min and then washed with buffer.
Samples are
extracted for 15 min with 50% ethanol and processed for LC/MS/MS to detect the
creatine
prodrug, creatine, and ATP levels. Increased creatine phosphate and ATP levels
in sodium
azide treated cells following exposure to a creatine prodrug indicates that
the prodrug is
capable of restoring cellular energy homeostasis.
Example 10: Protection Against 3-Nitropropionic Acid Induced Toxicity
[00323] An adaptation of the methods described by Brouillet et al., J.
Neurochem
2005, 95(6), 1521-40, is used to evaluate the protective effects on
intracellular energy
homeostasis of compounds of the invention.
[00324] The rat cardiomyoblast cell line H9c2 is obtained from ATCC (#CRL-
1446).
A 20 mM stock solution of 3-nitropropionic acid (3-NP) is prepared immediately
before use
in normal media (DMEM/High glucose (4.5 g/L)/10% FBS/6 mM L-glutamine/PSF) and
the
pH is adjusted to 7.4 by dropwi se addition of IN sodium hydroxide. A 40 mM
stock solution
of a creatine prodrug, e.g. a compound of the invention, is prepared in DMSO,
and creatine is
dissolved directly in serum-free media at 10 mM.
[00325] To measure the extent of cellular protection provided by the
creatine prodrug
and/or a creatine analog against 3-NP toxicity, H9c2 cells are plated in 96-
well clear-bottom
black tissue culture plates at 10K cells per well in normal media and
incubated overnight at
37 C. The following day the media is removed and replaced with serum-free
media
containing serial dilutions of a creatine prodrug or creatine. The plates are
incubated at 37 C
105

for 2 hours. Media is then removed by aspiration and replaced with normal
media containing various
concentrations of 3-NP and the plates incubated at 37 C for an additional 20
hours. To determine the
number of viable cells in each well, an equal volume of CellTiter-Glo reagent
(PromegaTm) is added and
mixed for 10 minutes on a plate shaker at room temperature. Luminescence is
measured by reading the
plates in a luminometer. The luminescence produced in this assay is
proportional to the amount of ATP
present, and directly relates to the number of metabolically active cells.
[00326] Increased viability of cells exposed to 3-NP and a creatine
prodrug compared to that of
cells exposed to 3-NP and creatine indicates that the creatine prodrug has the
capacity to maintain cellular
energy homeostasis.
Example 11: Pharmacokinetics of Creatine Prodrugs Following Colonic
Administration in Rats
[00327] Sustained release oral dosage forms, which release drug slowly
over periods of about 6 to
about 24 hours, generally release a significant proportion of the dose within
the colon. Thus, drugs
suitable for use in such dosage forms should be colonically absorbed. This
experiment is performed to
assess the uptake and resultant levels of a creatine prodrug and creatine in a
biological fluid such as the
plasma/blood or cerebrospinal fluid (CSF), following intracolonic
administration of a corresponding
creatine prodrug, such as a compound of the invention and thereby determine
the suitability of a creatine
prodrug for use in an oral sustained release dosage faun. Bioavailability of a
creatine prodrug and creatine
following co-administration of the creatine prodrug can be calculated relative
to oral administration
and/or to colonic administration of the creatine prodrug.
Step A: Administration Protocol
[00328] Rats are obtained commercially and are pre-cannulated in both the
ascending colon and
the jugular vein. Animals are conscious at the time of the experiment. All
animals are fasted overnight
and until 4 hours post-dosing of a creatine prodrug. The creatine prodrug is
administered as a solution
(in water or other appropriate solvent and vehicles) directly into the colon
via the cannula at a dose
equivalent to about 1 mg to about 200 mg of the creatine prodrug per kg body
weight. Blood samples
(0.3 mL) are obtained from the jugular cannula at intervals over 8 hours and
are immediately quenched
with sodium metabisulfite or other appropriate antioxidant to prevent
oxidation of the creatine prodrug.
Blood samples can be further quenched with methanol/perchloric acid to prevent
post-sampling
hydrolysis of the
106
Date Recue/Date Received 2022-04-14

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
creatine prodrug. Blood samples are analyzed as described below. Samples can
also be taken
from the CSF or other appropriate biological fluid.
Step B: Sample Preparation for Colonically Absorbed Prodrug
[00329] Methanol/perchloric acid (300 L) is added to blank 1.5 mL
Eppendorf tubes.
Rat blood (300 L) is collected into EDTA tubes containing 75 uL of sodium
metabisulfite at
different times and vortexed to mix. A fixed volume of blood (100 L) is
immediately added
into the Eppendorf tube and vortexed to mix. Ten microliters of a standard
stock solution of
the creatine prodrug (0.04, 0.2, 1, 5, 25, and 100 ug/mL) and 10 [IL of the
10% sodium
metabisulfite solution are added to 80 [11_, of blank rat blood to make up a
final calibration
standard (0.004, 0.02, 0.1, 0.5, 2.5, and 10 ug/mL). Methanol/perchloric acid
(300 [IL of
50/50) is then added into each tube followed by the addition of 20 uL of p-
chlorophenylalanine. The samples are vortexed and centrifuged at 14,000 rpm
for 10 min.
The supernatant is analyzed by LC/MS/MS.
Step C: LC/MS/MS Analysis
[00330] An API 4000 LC/MS/MS spectrometer equipped with Agilent 1100 binary
pumps, a CTC HTS-PAL autosampler, and a Zorbax XDB C8 4.6 x 150 mm column is
used
during the analysis. Appropriate mobile phases can be used such as, for
example, (A) 0.1%
formic acid, and (B) acetonitrile with 0.1% formic acid. Appropriate gradient
conditions can
be used such as, for example: 5% B for 0.5 min, then to 98% B in 3 min,
maintained at 98%
B for 2.5 min, and then returned to 2% B for 2 min. A TurboIonSpray source is
used on the
API 4000. The analysis is done in an appropriate ion mode and the MRM
transition for each
analyte is optimized using standard solution. 5 uL of each sample is injected.
Non-
compartmental analysis is performed using WinNonlin software (v.3.1
Professional Version,
Ph arsi ght Corporation, Mountain View, Calif) on individual animal profiles.
Summary
statistics on major parameter estimates is performed for Cmax (peak observed
concentration
following dosing), Tmax (time to maximum concentration is the time at which
the peak
concentration is observed), AUC(0.0 (area under the serum concentration-time
curve from
time zero to last collection time, estimated using the log-linear trapezoidal
method), AUC(0.
Jam) (area under the blood concentration time curve from time zero to
infinity, estimated
using the log-linear trapezoidal method to the last collection time with
extrapolation to
infinity), and t1/2,z (terminal half-life).
107

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
[00331] The pharmacokinetic parameters of the creatine prodrug and creatine
following colonic administration of the corresponding creatine prodrug are
determined and
compared to those obtained following an equivalent colonic dose of the
creatine prodrug.
Maximum concentrations of the creatine prodrug and creatine in the blood (Cmax
values) and
the area under blood concentration versus time curve (AUC) values after intra-
colonic dosing
of a creatine prodrug that are higher than those achieved for colonic
administration of the
corresponding creatine prodrug indicate that the prodrug provides enhanced
colonic
bioavailability.
Example 12: Pharmacokinetics of a Creatine Prodrug Following Intravenous or
Oral
Administration to Rats
[00332] A creatine prodrug is administered as an intravenous bolus
injection or by oral
gavage to groups of four to six adult male Sprague-Dawley rats (about 250 g).
Animals are
conscious at the time of the experiment. When orally administered, the
creatine prodrug is
administered as an aqueous solution (or as a solution of another appropriate
solvent
optionally including appropriate vehicles) at an appropriate creatine prodrug
dose equivalent
per kg body weight. Blood samples (0.3 mL) are obtained via a jugular vein
cannula at
intervals over 8 hours following oral dosing. Blood is quenched immediately
using, for
example, acetonitrile with 1% formic acid and then is frozen at 80 C until
analyzed.
Samples may also be taken form the CSF or other appropriate biological fluid.
[00333] Three hundred (300) iL of 0.1% formic acid in acetonitrile is added
to blank
1.5 mL tubes. Rat blood (300 [IL) is collected at different times into tubes
containing EDTA
and vortexed to mix. A fixed volume of blood (100 L) is immediately added
into the tube
and vortexed to mix. Ten microliters of a creatine prodrug standard stock
solution (0.04, 0.2,
1, 5, 25, and 100 mg/mL) is added to 90 L of blank rat blood quenched with
300 .1_, of 0.1%
fottnic acid in acetonitrile. Then, 20 L of p-chlorophenylalanine is added to
each tube to
make a final calibration standard (0.004, 0.02, 0.1, 05, 2.5, and 10 jig/mL).
Samples are
vortexed and centrifuged at 14,000 rpm for 10 min. The supernatant is analyzed
by
LC/MS/MS.
[00334] An API 4000 LC/MS/MS spectrometer equipped with Agilent 1100 binary
pumps, a CTC HTS-PAL autosampler, and a Phenomenex Synergihydro-RP 4.6 x 30 mm
column are used in the analysis. Appropriate mobile phases and gradient
conditions are used
for the analysis. The analysis is done in the appropriate ion mode and the MRM
transition for
each analyte is optimized using standard solutions. Five (5) L of each sample
is injected.
108

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
Non-compartmental analysis is performed using WinNonlin (v.3.1 Professional
Version,
Pharsight Corporation, Mountain View, Calif.) on individual animal profiles.
Summary
statistics on major parameter estimates is performed for Cma, (peak observed
concentration
following dosing), Tmax (time to maximum concentration is the time at which
the peak
concentration was observed), AUC(0t) (area under the serum concentration-time
curve from
time zero to last collection time, estimated using the log-linear trapezoidal
method), AUC(0.
win), (area under the serum concentration time curve from time zero to
infinity, estimated
using the log-linear trapezoidal method to the last collection time with
extrapolation to
infinity), and tin. (terminal half-life).
[00335] The oral bioavailability (F(%) of the creatine prodrug is
deteimined by
comparing the area under the creatine prodrug concentration vs time curve
(AUC) following
oral administration with the AUC of the creatine prodrug concentration vs time
curve
following intravenous administration of the creatine prodrug on a dose
normalized basis.
[00336] Samples can also be obtained from the CSF and the pharmacokinetics
of the
creatine prodrug and creatine determined. Higher levels of creatine prodrug
and/or creatine
can indicate that the prodrug has the ability to be translocated across the
blood-brain barrier.
[00337] Similar studies on the pharmacokinetics of a creatine prodrug can
be
performed in other animals including but not limited to dogs, monkeys, and
human.
Example 13: Use of Animal Models to Assess the Efficacy of Creatine Prodrugs
for Treating
Amyotrophic Lateral Sclerosis
[00338] A murine model of SOD1 mutation-associated ALS has been developed
in
which mice express the human superoxide dismutase (SOD) mutation
glycine.fwdarw.alanine
at residue 93 (SOD1). These SOD' mice exhibit a dominant gain of the adverse
property of
SOD, and develop motor neuron degeneration and dysfunction similar to that of
human ALS
(Gurney et al., Science 1994, 264(5166), 1772-1775; Gurney et al., Ann Neurol.
1996, 39,
147-157; Gurney, J. Neurol. Sci. 1997, 152, S67-73; Ripps et al., Proc Natl
Acad Sci U.S.A.
1995, 92(3), 689-693; and Bruijn et al., Proc Natl Acad Sci U.S.A. 1997,
94(14), 7606-7611).
The SOD1 transgenic mice show signs of posterior limb weakness at about 3
months of age
and die at 4 months. Features common to human ALS include astrocytosis,
microgliosis,
oxidative stress, increased levels of cyclooxygenase/prostaglandin, and as the
disease
progresses, profound motor neuron loss.
[00339] Studies are performed on transgenic mice overexpressing human Cu/Zn-
SOD
G93A mutations (B6SJL-TgN(SOD1-G93A) 1 Gur) and non-transgenic B6/SJL mice and
109

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
their wild litter mates. Mice are housed on a 12-hr day/light cycle and
(beginning at 45 d of
age) allowed ad libitum access to either test compound-supplemented chow or as
a control,
regular formula cold press chow processed into identical pellets. Genotyping
can be
conducted at 21 days of age as described in Gurney et al., Science 1994,
264(5166), 1772-
1775. The SOD1 mice are separated into groups and treated with a test compound
or serve as
controls.
[00340] The mice are observed daily and weighed weekly. To assess health
status mice
are weighed weekly and examined for changes in lacrimation/salivation,
palpebral closure,
ear twitch and pupillary responses, whisker orienting, postural and righting
reflexes and
overall body condition score. A general pathological examination is conducted
at the time of
sacrifice.
[00341] Motor coordination performance of the animals can be assessed by
one or
more methods known to those skilled in the art. For example, motor
coordination can be
assessed using a neurological scoring method. In neurological scoring, the
neurological score
of each limb is monitored and recorded according to a defined 4-point scale:
0=normal reflex
on the hind limbs (animal splays its hind limbs when lifted by its tail);
1=abnormal reflex of
hind limbs (lack of splaying of hind limbs when animal is lifted by the tail);
2=abnormal
reflex of limbs and evidence of paralysis; 3=lack of reflex and complete
paralysis; and
4=inability to right when placed on the side in 30 seconds or found dead. The
primary end
point is survival with secondary end points of neurological score and body
weight.
Neurological score observations and body weight are made and recorded five
days per week.
Data analysis is performed using appropriate statistical methods.
[00342] The rotarod test evaluates the ability of an animal to stay on a
rotating dowel
allowing evaluation of motor coordination and proprioceptive sensitivity. The
apparatus is a 3
cm diameter automated rod turning at, for example, 12 rounds per min. The
rotarod test
measures how long the mouse can maintain itself on the axle without falling.
The test can be
stopped after an arbitrary limit of, for example, 120 sec. If the animal falls
before 120 sec, the
performance is recorded and two additional trials are performed. The mean time
of 3 trials is
calculated. A motor deficit is indicated by a decrease of walking time.
[00343] In the grid test, mice are placed on a grid (length: 37 cm, width:
10.5 cm, mesh
size: 1 x 1 cm2) situated above a plane support. The number of times the mice
put their paws
through the grid is counted and serves as a measure for motor coordination.
[00344] The hanging test evaluates the ability of the animal to hang on a
wire. The
apparatus is a wire stretched horizontally 40 cm above a table. The animal is
attached to the
110

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
wire by its forepaws. The time needed by the animal to catch the string with
its hind paws is
recorded (60 sec max) during three consecutive trials.
[00345] Electrophysiological measurements (EMG) can also be used to assess
motor
activity condition. Electromyographic recordings are performed using an
electromyography
apparatus. During EMG monitoring the mice are anesthetized. The measured
parameters are
the amplitude and the latency of the compound muscle action potential (CMAP).
CMAP is
measured in gastrocnemius muscle after stimulation of the sciatic nerve. A
reference
electrode is inserted near the Achilles tendon and an active needle placed at
the base of the
tail. A ground needle is inserted on the lower back of the mice. The sciatic
nerve is stimulated
with a single 0.2 msec pulse at supramaximal intensity (12.9 mA). The
amplitude (mV) and
the latency of the response (ms) are measured. The amplitude is indicative of
the number of
active motor units, while distal latency reflects motor nerve conduction
velocity.
[00346] The efficacy of test compounds can also be evaluated using
biomarker
analysis. To assess the regulation of protein biomarkers in SOD1 mice during
the onset of
motor impairment, samples of lumbar spinal cord (protein extracts) are applied
to
ProteinChip Arrays with varying surface chemical/biochemical properties and
analyzed, for
example, by surface enhanced laser desorption ionization time of flight mass
spectrometry.
Then, using integrated protein mass profile analysis methods, data is used to
compare protein
expression profiles of the various treatment groups. Analysis can be performed
using
appropriate statistical methods.
Example 14: Clinical Trials to Assess the Efficacy of Creatine Prodrugs for
Treating
Parkinson's Disease
[00347] The following clinical study may be used to assess the efficacy of
a creatine
prodrug in treating Parkinson's disease. Patients with idiopathic PD
fulfilling the Queen
Square Brain Bank criteria (Gibb et al., J Neurol Neurosurg Psychiatry 1988,
51, 745-752)
with motor fluctuations and a defined short duration GABA analog response (1.5-
4 hours) are
eligible for inclusion. Clinically relevant peak dose dyskinesias following
each morning dose
of their current medication are a further pre-requisite. Patients are also
required to have been
stable on a fixed dose of treatment for a period of at least one month prior
to starting the
study. Patients are excluded if their current drug regime includes slow-
release formulations of
L-Dopa, COMT inhibitors, selegiline, anticholinergic drugs or other drugs that
could
potentially interfere with gastric absorption (e.g. antacids). Other exclusion
criteria include
patients with psychotic symptoms or those on antipsychotic treatment patients
with clinically
111

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
relevant cognitive impailment, defined as MIMS (Mini Mental State) score of
less than 24
(Folstein et al., J Psychiatr Res 1975, 12, 189-198), risk of pregnancy, Hoehn
& Yahr stage 5
in off-status, severe, unstable diabetes mellitus, and medical conditions such
as unstable
cardiovascular disease or moderate to severe renal or hepatic impairment. Full
blood count,
liver, and renal function blood tests are taken at baseline and after
completion of the study.
[00348] A randomized, double-blind, and cross-over study design is used.
The
pharmacokinetics of a creatine prodrug and released creatine can be assessed
by determining
the blood concentrations at appropriate time intervals. Creatine levels in the
brain can also be
determined non-invasively by magnetic resonance spectroscopy (MRS).
[00349] For clinical assessment, motor function is assessed using UPDRS
(United
Parkinson's Disease Rating Scale) motor score and BrainTest (Giovanni et al.,
J Neural
Neurosurg Psychiatry 1999, 67, 624-629), which is a tapping test performed
with the patient's
more affected hand on the keyboard of a laptop computer. These tests are
carried out at
baseline and then immediately following each blood sample until patients reach
their full on-
stage, and thereafter at intervals until patients reach their baseline off-
status. Once patients
reach their full on-state, video recordings are performed three times at 20
min intervals. The
following mental and motor tasks, which have been shown to increase dyskinesia
(Duriff et
al., Mov Disord 1999, 14, 242-245) are monitored during each video session:
(1) sitting still
for 1 minute; (2) performing mental calculations; (3) putting on and buttoning
a coat; (4)
picking up and drinking from a cup of water; and (5) walking. Videotapes are
scored using,
for example, versions of the Goetz Rating Scale and the Abnormal Involuntary
Movements
Scale to document a possible increase in test compound induced dyskinesia.
[00350] Actual occurrence and severity of dyskinesia is measured with a
Dyskinesia
Monitor (Manson et al., J Neural Neurosurg Psychiatry 2000, 68, 196-201). The
device is
taped to a patient's shoulder on their more affected side. The monitor records
during the entire
time of a challenging session and provides a measure of the frequency and
severity of
occurring dy ski nesi as.
[00351] Results can be analyzed using appropriate statistical methods.
Example 15: Efficacy of Creatine Prodrugs in MPTP Induced Neurotoxicity Animal
Model
of Parkinson's Disease
[00352] MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) is a neurotoxin
that
produces a Parkinsonian syndrome in both man and experimental animals. Studies
of the
mechanism of MPTP neurotoxicity show that it involves the generation of a
major
112

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
metabolite, MPP+, formed by the activity of monoamine oxidase on MPTP.
Inhibitors of
monoamine oxidase block the neurotoxicity of MPTP in both mice and primates.
The
specificity of the neurotoxic effects of MPP+ for dopaminergic neurons appears
to be due to
the uptake of MPP- by the synaptic dopamine transporter. Blockers of this
transporter prevent
MPP+neurotoxicity. MPP+ has been shown to be a relatively specific inhibitor
of
mitochondrial complex I activity, binding to complex I at the retenone binding
site and
impairing oxidative phosphorylation. In vivo studies have shown that MPTP can
deplete
striatal ATP concentrations in mice. It has been demonstrated that MPP+
administered intra-
striatally in rats produces significant depletion of ATP as well as increased
lactate
concentration confined to the striatum at the site of the injections.
Compounds that enhance
ATP production can protect against MPTP toxicity in mice.
[00353] A creatine prodrug is administered to animals such as mice or rats
for three
weeks before treatment with MPTP. MPTP is administered at an appropriate dose,
dosing
interval, and mode of administration for 1 week before sacrifice. Control
groups receive
either normal saline or MPTP hydrochloride alone. Following sacrifice the two
striate are
rapidly dissected and placed in chilled 0.1 M perchloric acid. Tissue is
subsequently
sonicated and aliquots analyzed for protein content using a fluorometer assay.
Dopamine,
3,4-dihydroxyphenylacetic acid (DOPAC), and homovanillic acid (HVA) are also
quantified.
Concentrations of dopamine and metabolites are expressed as nmol/mg protein.
[00354] Creatine prodrugs that protect against DOPAC depletion induced by
MPTP,
HVA, and/or dopamine depletion are neuroprotective and therefore can be useful
for the
treatment of Parkinson's disease.
Example 16: Evaluation of Potential Anti-Parkinsonian Activity Using a
Haloperidol-
Induced Hypolocomotion Animal Model
[00355] It has been demonstrated that adenosine antagonists, such as
theophylline, can
reverse the behavioral depressant effects of dopamine antagonists, such as
haloperidol, in
rodents and is considered a valid method for screening drugs with potential
antiparkinsonian
effects (Mandhane, et al., Eur. J. Pharmacol. 1997, 328, 135-141). The ability
of creatine
prodrugs to block haloperidol-induced deficits in locomotor activity in mice
can be used to
assess both in vivo and potential anti-Parkinsonian efficacy.
[00356] Mice used in the experiments are housed in a controlled environment
and
allowed to acclimatize before experimental use. 1.5 h before testing, mice are
administered
0.2 mg/kg haloperidol, a dose that reduces baseline locomotor activity by at
least 50%. A test
113

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
compound is administered 5-60 min prior to testing. The animals are then
placed individually
into clean, clear polycarbonate cages with a flat perforated lid. Horizontal
locomotor activity
is determined by placing the cages within a frame containing a 3 x 6 array of
photocells
interfaced to a computer used to tabulate beam interrupts. Mice are left
undisturbed to
explore for 1 h, and the number of beam interruptions made during this period
serves as an
indicator of locomotor activity, which is compared with data for control
animals for
statistically significant differences.
Example 17: 6-Hydroxydopamine Animal Model of Parkinson's Disease
[00357] The neurochemical deficits seen in Parkinson's disease can be
reproduced by
local injection of the dopaminergic neurotoxin, 6-hydroxydopamine (6-0HDA)
into brain
regions containing either the cell bodies or axonal fibers of the
nigrostriatal neurons. By
unilaterally lesioning the nigrostriatal pathway on only one-side of the
brain, a behavioral
asymmetry in movement inhibition is observed. Although unilaterally-lesioned
animals are
still mobile and capable of self maintenance, the remaining dopamine-sensitive
neurons on
the lesioned side become supersensitive to stimulation. This is demonstrated
by the
observation that following systemic administration of dopamine agonists, such
as
apomorphine, animals show a pronounced rotation in a direction contralateral
to the side of
lesioning. The ability of compounds to induce contralateral rotations in 6-
0HDA lesioned
rats has been shown to be a sensitive model to predict drug efficacy in the
treatment of
Parkinson's disease.
[00358] Male Sprague-Dawley rats are housed in a controlled environment and
allowed to acclimatize before experimental use. Fifteen minutes prior to
surgery, animals are
given an intraperitoneal injection of the noradrenergic uptake inhibitor
desipramine (25
mg/kg) to prevent damage to nondopamine neurons. Animals are then placed in an
anaesthetic chamber and anaesthetized using a mixture of oxygen and
isoflurane. Once
unconscious, the animals are transferred to a stereotaxic frame, where
anesthesia is
maintained through a mask. The top of the animal's head is shaved and
sterilized using an
iodine solution. Once dry, a 2 cm long incision is made along the midline of
the scalp and the
skin retracted and clipped back to expose the skull. A small hole is then
drilled through the
skull above the injection site. In order to lesion the nigrostriatal pathway,
the injection
cannula is slowly lowered to position above the right medial forebrain bundle
at -3.2 mm
anterior posterior, -1.5 mm medial lateral from the bregma, and to a depth of
7.2 mm below
the duramater. Two minutes after lowering the cannula, 6-0HDA is infused at a
rate of 0.5
114

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
[IL/min over 4 min, yielding a final dose of 8 lug. The cannula is left in
place for an additional
min to facilitate diffusion before being slowly withdrawn. The skin is then
sutured shut, the
animal removed from the stereotaxic frame, and returned to its housing. The
rats are allowed
to recover from surgery for two weeks before behavioral testing.
[00359] Rotational behavior is measured using a rotometer system having
stainless
steel bowls (45 cm diameter x.15 cm high) enclosed in a transparent Plexiglas
cover running
around the edge of the bowl and extending to a height of 29 cm. To assess
rotation, rats are
placed in a cloth jacket attached to a spring tether connected to an optical
rotometer
positioned above the bowl, which assesses movement to the left or right either
as partial (450)
or full (3600 rotations).
[00360] To reduce stress during administration of a test compound, rats are
initially
habituated to the apparatus for 15 min on four consecutive days. On the test
day, rats are
given a test compound, e.g., a creatine prodrug. Immediately prior to testing,
animals are
given a subcutaneous injection of a subthreshold dose of apomorphine, and then
placed in the
harness and the number of rotations recorded for one hour. The total number of
full
contralateral rotations during the hour test period serves as an index of
antiparkinsonian drug
efficacy.
Example 18: Animal Studies to Assess the Efficacy of Creatine Prodrugs in
Ischemic Injury
[00361] Adult male rats are given a creatine prodrug and, after about 24 h,
are
anesthetized and prepared for coronary artery occlusion. An additional dose of
a creatine
prodrug is administered at the start of the procedure and the left main
coronary artery
occluded for 30 min and then released. The same dose of a creatine prodrug is
then
administered at appropriate intervals and duration following surgery. The
animals are then
studied for cardiac function. Animals receiving a sham injection (saline)
demonstrate a large
increase in the left end diastolic pressure, indicative of a dilated, stiff
heart secondary to
myocardial infarction Creatine prodrugs that eliminate or reduce the deficit
in cardiac
function compared to sham operated control are useful in preventing ischemic
injury.
Example 19: Animal Studies to Assess the Ability of Creatine Prodrugs to
Maintain Organ
Viability
[00362] Wistar male rats weighing 300 to 330 g are administered a creatine
prodrug or
vehicle 24 h prior to removal of the heart for ex vivo studies. Animals are
sacrificed with
pentobarbital (0.3 mL) and intravenously heparinized (0.2 mL). The hearts are
initially
115

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
allowed to equilibrate for 15 min. The left ventricular balloon is then
inflated to a volume that
gives an end-diastolic pressure of about 8 mm Hg. A left ventricular pressure-
volume curve is
constructed by incremental inflation of the balloon volume by 0.02 mL
aliquots. Zero volume
is defined as the point at which the left ventricular end-diastolic pressure
is zero. On
completion of the pressure-volume curve, the left ventricular balloon is
deflated to set end-
diastolic pressure back to 8 mmHg and the control period is continued for 15
min after check
of coronary flow. The heart is then arrested with 50 mL Celsior + molecule to
rest at 4 C
under a pressure of 60 cm H20. The heart is then removed and stored for 5 h at
4 C in a
plastic container filled with the same solution and surrounded with crushed
ice.
[00363] After storage, the heart is transferred to a Langendorff apparatus.
The balloon
catheter is re-inserted into the left ventricle and re-inflated to the same
volume as during the
preischemic period. The heart is reperfused for at least 2 h at 37 C. The re-
perfusion
pressure is set at 50 cm H20 for 15 min of re-flow and then back to 100 cm H20
for the 2
next h. Pacing (320 beats per min) is re-instituted. Isovolumetric
measurements of contractile
indexes and diastolic pressure are taken in triplicate at 25, 45, 60, and 120
min of reperfusion.
At this time point pressure volume curves are obtained and coronary effluent
during the 45
min reperfusion collected to measure creatine kinase leakage. Improved left
ventricular
pressure following treatment with a prodrug of a creatine analog, as well as
improved
volume-pressure curve, decrease of left diastolic ventricular pressure and
decrease of creatine
kinase leakage indicates the ability of the creatine prodrug to maintain organ
viability.
Example 20: Neuroprotective Effects of Prodrugs of Creatine Analogs in a
Transgenic
Mouse Model of Huntington's Disease
[00364] Transgenic HD mice of the N171-82Q strain and non-transgenic
littermates
are treated with a prodrug of a creatine analog or a vehicle from 10 weeks of
age. The mice
are placed on a rotating rod ("rotarod"). The length of time at which a mouse
falls from the
rotarod is recorded as a measure of motor coordination. The total distance
traveled by a
mouse is also recorded as a measure of overall locomotion. Mice administered
creatine
prodrugs that are neuroprotective in the N171-82Q transgenic HD mouse model
remain on
the rotarod for a longer period of time and travel further than mice
administered vehicle.
Example 21: Efficacy of Creatine Prodrugs in a Malonate Model of Huntington's
Disease
[00365] A series of reversible and irreversible inhibitors of enzymes
involved in
energy generating pathways has been used to generate animal models for
neurodegenerative
116

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
diseases such as Parkinson's and Huntington's diseases. Inhibitors of
succinate
dehydrogenase, an enzyme that impacts cellular energy homeostasis, has been
used to
generate a model for Huntington's disease (Brouillet et al., J. Neurochem.
1993, 60, 356-359;
Beal et al., J. Neurosci. 1993, 13, 4181-4192; Henshaw et al., Brain Research
1994, 647, 161-
166 (1994); and Beal et al., J. Neurochem. 1993, 61, 1147-1150). The enzyme
succinate
dehydrogenase plays a central role in both the tricarboxylic acid cycle as
well as the electron
transport chain in the mitochondria. Malonate is a reversible inhibitor
malonate of succinate
dehydrogenase. Intrastriatal injections of malonate in rats have been shown to
produce dose
dependent striatal excitotoxic lesions that are attenuated by both competitive
and
noncompetitive NMDA antagonists (Henshaw et al., Brain Research 1994, 647, 161-
166).
The glutamate release inhibitor, lamotrigine, also attenuates the lesions. Co-
injection with
succinate blocks the lesions, consistent with an effect on succinate
dehydrogenase. The
lesions are accompanied by a significant reduction in ATP levels as well as
significant
increase in lactate levels in vivo as shown by chemical shift resonance
imaging (Beal et al., J.
Neurochem. 1993, 61, 1147-1150). The lesions produced the same pattern of
cellular sparing,
which is seen in Huntington's disease, supporting malonate challenge as a
useful model for
the neuropathologic and neurochemical features of Huntington's disease.
[00366] To evaluate the effect of creatine prodrugs in this malonate model
for
Huntington's disease, a creatine prodrug is administered at an appropriate
dose, dosing
interval, and route, to male Sprague-Dawley rats. A prodrug is administered
for two weeks
prior to the administration of malonate and then for an additional week prior
to sacrifice.
Malonate is dissolved in distilled deionized water and the pH adjusted to 7.4
with 0.1 M HC1.
Intrastriatal injections of 1.5 [IL of malonate containing 3 [tmol are made
into the left
striatum at the level of the Bregma 2.4 mm lateral to the midline and 4.5 mm
ventral to the
dura. Animals are sacrificed at 7 days by decapitation and the brains quickly
removed and
placed in ice cold 0.9% saline solution. Brains are sectioned at 2 mm
intervals in a brain
mold. Slices are then placed posterior side down in 2% 2,3,5-
triphenyltetrazolium chloride.
Slices are stained in the dark at room temperature for 30 min and then removed
and placed in
4% paraformaldehyde pH 7.3. Lesions, noted by pale staining, are evaluated on
the posterior
surface of each section. The measurements are validated by comparison with
measurements
obtained on adjacent Nissl stain sections.
[00367] Compounds exhibiting a neuroprotective effect and therefore useful
in treating
Huntington's disease show a reduction in malonate-induced lesions.
117

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
Example 22: Efficacy of Creatine Prodrugs in a Model of Creatine Transporter
Disorder
[00368] A mouse model of human CrT deficiency has been generated, which
enables
development of treatments for this condition (Skelton et al., PloS One, 201, 6
(1), e16187).
Mice with exons 2-4 of Slc6a8 flanked by loxP sites were crossed to Cre:CMV
mice to
create a line of ubiquitous CrT knockout expressing mice. Male CrT¨/y
(affected) mice lack
Cr in the brain and muscle with significant reductions of Cr in other tissues
including heart
and testes. CrT¨/y mice show increased path length during acquisition and
reversal learning
in the Morris water maze. During probe trials, CrT¨/y mice show increased
average distance
from the platform site. CrT¨/y mice show reduced novel object recognition and
conditioned
fear memory compared to CrT+/y. CrT¨/y mice have increased serotonin and 5-
hydroxyindole acetic acid in the hippocampus and prefrontal cortex. Ubiquitous
CrT
knockout mice have learning and memory deficits resembling human CrT
deficiency and this
model is useful in understanding this disorder and testing creatine prodrugs
as therapies for
this disorder.
[00369] To evaluate the effect of creatine prodrugs in the Morris Water
Maze (MWM),
a creatine prodrug is administered at an appropriate dose, dosing interval,
and route, to male
CrT¨/y mice. The MWM is a test of spatial learning and reference memory
(Vorhees et al.,
Nature Protocols 2006, 1: 848-858). and animals are tested as described by
Skelton et al.,
Brain Res 2003, 984: 1-101 and Schaefer et al., Neuroscience 2009, 164: 1431-
1443. Prior
to hidden platform testing, visible platform training (cued learning) is
conducted for 6 days.
During this phase, curtains are closed around the maze to obscure prominent
distal cues and a
cm diameter platform with an orange ball mounted above it on a brass rod is
placed in a
predetermined quadrant. On the first day, 6 trials (90 s) are administered
with the platform
and start in the same position; 2 trials per day are given on subsequent days
with the start and
platform positions randomized.
[00370] The hidden platform portion of the MWM test is conducted in three
phases (6
days/phase: acquisition, reversal, and shift) consisting of 4 trials per day
for 6 days for
animals to learn the location of the hidden platform followed by a single
probe trial (no
platform) on day 7 (Vorhees et al., Nature Protocols 2006, 1. 848-858).
Platform diameters
are 10 cm for acquisition, 7 cm for reversal (located in the opposite
quadrant), and 5 cm for
shift, (located in one of the adjacent quadrants). Performance is measured
using AnyMaze
software (Stoelting Company, Wood Dale, IL). The effect of prodrug treatment
is analyzed
by comparing performance of control (untreated male CrT¨/y mice and/or wild
type mice) to
prodrug treated mice.
118

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
[00371] To evaluate the effect of creatine prodrugs in the Conditioned Fear
model, a
creatine prodrug is administered at an appropriate dose, dosing interval, and
route, to male
CrT¨/y mice. Cued and contextual fear is assessed as described by Peters et
al., Science
2010, 328: 1288-1290). On day 1, untreated (control) and treated (prodrug
administered)
mice are exposed to 30 tones (82 dB, 2 kHz, 30 s on/off cycle) followed by 3
tone-footshock
pairings (0.5 mA for 1 s). On the following day, animals are returned to the
chamber with no
tone or shock presented as a test of contextual fear. The next day, animals
are placed in the
chamber with a novel grid floor. Following 3 min acclimatization, the tone is
presented and
freezing behavior scored. Animals are then exposed to 30 cycles of 30 s with
and 30 s
without tone to measure fear extinction. Freezeframe software and Coulbourn
test chambers
are used (Coulboum Instruments, Allentown, PA). Percent time freezing is
analyzed. The
effect of prodrug treatment is analyzed by comparing performance of control
(untreated male
CrT¨/y mice and/or wild type mice) to prodrug treated mice.
[00372] To evaluate the effect of creatine prodrugs in the Novel Object
Recognition
(NOR) model, a creatine prodrug is administered at an appropriate dose, dosing
interval, and
route, to male CrT¨/y mice. NOR is a test of short-term memory (Clark et al.,
J Neurosci
2000, 20: 8853-8860). Mice are habituated to the arena (91 cm diameter) for 2
days (10
min/day) followed by 2 days (10 min/day) of habituation to two identical
objects. On the test
day, animals are presented with two new identical objects until 30 s of
cumulative
observation time is obtained. One hour later memory is tested by presenting
the animal with
an identical copy of one of the familiar objects along with a novel object. A
discrimination
index is calculated by subtracting the time observing the familiar object from
time spent
observing the novel object. The effect of prodrug treatment is analyzed by
comparing
performance of control (untreated male CrT¨/y mice and/or wild type mice) to
prodrug
treated mice.
[00373] Compounds useful in treating creatine transporter disorder will
show an
improvement in treated male CrT-/Y mice relative to untreated controls in one
or more of the
evaluations outlined above or in alternative models for testing behavior,
neurological and/or
neuromuscular function.
119

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
Example 23: Synthesis of ethyl (N' -hy droxy-N-m ethyl carb amimi dami
do)acetate
HO....
NH2OH. HC1
C2H5 TEA. Et0H
H2N/'N\ N ()' ...,õ
Reflux 21 15
CH3 0
CH3 0
[00374] Ethyl (N'-hydroxy-N-methylcarbamimidamido) acetate can be
synthesized
using the procedure described in Zbinden et al., Bioorganic & Medicinal
Chemistry Letters,
2005, 15: 5344-5352. Briefly, ethyl [cyano(methyl)amino] acetate, available
from MP
Biomedicals, Inc., is reflux with hydroxylamine hydrochloride in Et0H to give
the title
compound.
Example 24: Synthesis of tert-butyl 2-(3-(tert-butoxycarbony1)-2-hydroxy-1-
methylguanidino)acetate -3-yl)amino]acetate
Step 1 Step 2 N,OH
HC1 Br, K2CO3, ACN). NH2OH, THF 11
1
HN n
overnight 1 RT, 30 min 8 0 I 0
Step 3
N-OH
Boc20, THF 11
RT Boc,, overnight N N n
H 8
Step 1: Synthesis of tert-butyl 2-(N-methylcyanamido)acetate
NC,
N
1 8
[00375] A round bottom flask equipped with a stir bar was charged with tert-
butyl 2-
(methylamino)acetate (500 mg, 2.75 mmol), potassium carbonate (761 mg, 5.50
mmol) and
acetonitrile (10 mL). The mixture was stirred at room temperature for 30 min
at which time a
solution of cyanogen bromide (320 mg, 3.025 mmol) in acetonitrile (2 ml) was
added. The
reaction was allowed to stir overnight at room temperature and the solvent was
decanted
leaving behind an insoluble residue. The solvent was then evaporated under
reduced pressure
and the material was purified by flash chromatography eluting with petroleum
ether ¨ ethyl
acetate (gradient 0% to 40% ethyl acetate) to give tert-butyl 2-(N-
methylcyanamido)acetate
120

CA 02971729 2017-06-20
WO 2016/106284
PCT/1JS2015/067283
(410 mg, 2.41 mmol, 88% yield) as a white solid. ES LC-MS m/z = 171 (1\4+H+) &
193
(M+Na+).
Step 2: Synthesis of tert-butyl 2-(2-hydroxy-1-methylguanidino)acetate
NõOH
H2N
1 8
[00376] A round
bottom flask equipped with a stir bar was charged with tert-butyl 2-
(N-methylcyanamido)acetate (300 mg, 1.76 mmol) and tetrahydrofuran (5 mL). To
this
mixture was added hydroxylamine (50% aqueous, 583 mg, 8.80 mmol). After 1
hour, 10 mL
of water was added and the mixture was extracted with 5 ml dichloromethane 3
times. The
organic layer was then dried with MgSO4, filtered and concentrated under
reduced pressure to
give crude tert-butyl 2-(2-hydroxy-1-methylguanidino)acetate (349 mg, 1.72
mmol, 98%
yield) as a white solid which was used immediately in the next step. ES LC-MS
m/z = 204
(M+H+).
Step 3: Synthesis of tert-butyl 2-(3-(tert-butoxycarbony1)-2-hydroxy-1-
methylguanidino)acetate -3-yl)amino]acetate
_OH
Boc,
N
H I 8
[00377] A round
bottom flask equipped with a stir bar was charged with tert-butyl 2-
(2-hydroxy-1-methylguanidino)acetate (349 mg, 1.72 mmol) and tetrahydrofuran
(5 mL). To
this mixture was added di-tert-butyl dicarbonate (375 mg, 1.72 mmol). The
reaction was
stirred at room temperature overnight. The solvent was then evaporated under
reduced
pressure and the material was purified by flash chromatography eluting with
dichloromethane
- ethyl acetate (gradient 0% to 30% ethyl acetate) to give tert-butyl 2-(3-
(tert-
butoxycarbony1)-2-hydroxy-l-methylguanidino)acetate (151 mg, 0.50 mmol, 29%
yield) as a
white solid. ES LC-MS m/z = 304 (M+H+). NMR (DIMETHYL SULFOXIDE-d) 6: 5.73
(s, 2H), 3.81 (s, 2H), 2.76 (s, 3H), 1.42 (s, 9H), 1.41 (s, 9H).
Example 25: Synthesis of tert-butyl 2-(3-(tert-butoxycarbony1)-2-hydroxy-1-
trideuteriomethylguanidino)acetate
121

CA 02971729 2017-06-20
WO 2016/106284
PCT/1JS2015/067283
Step I
HCI
riii SO2CI Step 2
0õ0 Rõp
H2N'-'( 02N ''' 0 ,s,..y0, 0,,,
..,
0 H '
pyridine. 0 C, 0 110 0,3 0
Cs2CO3, DMF 02N
02N
rt, 90 min
Step 3
,k I Step 4 Step 5
i) PhSH, Cs2CO3 TFA
acetonitrile, THF/10:1, 45 C õ,.,,
________________ ..- o yThr'
TFA 30%, DCM ,..., FiNii,-(3-1/ BrCN, K2CO3, ACN
____________________________________________________________________ ).
ii) BOC20. H20/CH2C12 CD3 0 ' 0 C, 3h 0D3 0 RT,
overnight
Step 6
N_OH Step 7
N_ON
NC0 NH2OH, THF ,I1 L ot, RBoc20, THFovernight II
" H2N f\l _______ N.- Boc,N,AN-y0,,..-
D3 0 RT, 30 min
CD3 0 H 1
CD3 0
Step 1: Synthesis of tert-butyl 2-(4-nitrophenylsulfonamido) acetate
0õp
0 \S.N....--).(0.,.....õ--
H
02N 0
[00378] A round bottom flask equipped with a stir bar and under nitrogen
was charged
with glycine tert-butyl ester hydrochloride (20 g, 119.76 mmol) and pyridine
(260 mL). The
mixture was cooled to 0 C at which time 4-nitrobenzenesulfonyl chloride
(28.98 g, 131.74
mmol) was added portion-wise maintaining the mixture below 10 C. The reaction
was then
allowed to warm to room temperature. After 18 h at room temperature, the
reaction mixture
was poured into water (1000 mL). A precipitate formed which was filtered and
dried under
vacuum to give tert-butyl 2-(4-nitrophenylsulfonamido) acetate (30.8 g, 97.46
mmol, 81%
yield) as a yellow solid. 111 NMR (CDC13) 6: 8.36-8.34 (m, 2H), 8.07-8.05 (m,
2H), 5.23 (br
s, 1H), 3.75-3.74 (d, ,I= 5.6 Hz, 2H), 1.35 (s, 9H).
Step 2: Synthesis of tert-butyl 2-[(4-nitrophenyl)sulfonyl-
(trideuteriomethyl)amino]acetate
0õp
0
02N
1
CD3 0
[00379] A round bottom flask equipped with a stir bar and under nitrogen
was charged
with tert-butyl 2-(4-nitrophenylsulfonamido) acetate (30.8 g, 97.46 mmol), DMF
(320 mL)
and CD3I (14.13 g, 97.46 mmol). To this mixture at room temperature was added
Cs2CO3
(34.85 g, 107.22 mmol) and the reaction was stirred for 45 min. The reaction
mixture was
122

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
then poured into water (1000 mL) and extracted with Et0Ac (3 x 500 mL). The
combined
organic phases were washed with NaCl (500 mL), dried (Na2SO4), filtered and
the solvent
was evaporated under reduced pressure to give tert-butyl 244-
nitrophenyl)sulfonyl-
(trideuteriomethyl)amino[acetate (27.8 g, 83.48 mmol, 81% yield as a pale
yellow solid. 1H
NMR (CDC13) 3: 8.36-8.33 (d, J = 8.8 Hz, 2H), 8.01-7.99 (d, J= 9.2 Hz, 2H),
3.98 (s, 2H),
1.38 (s, 9H).
Step 3: Synthesis of tert-butyl 2-(tert-
butoxycarbonyl(trideuteriomethyl)amino)acetate
1/ I
CD3 0
[00380] A round bottom flask equipped with a stir bar and under nitrogen
was charged
tert-butyl 2-[(4-nitrophenyl)sulfonyktrideuteriomethyl)amino]acetate (27.8 g,
83.48 mmol),
Cs2CO3 (67.83 g, 208.7 mmol) acetonitrile (400 mL) and THE (40 mL). To this
solution was
added thiophenol (34 mL, 333.93 mmol) and the reaction was heated at 45 C for
90 min at
which time the reaction mixture was diluted with MTBE (500 mL) and extracted
with water
(5 x 100 mL). The combined water extracts were washed with MTBE (500 mL) and
to the
water mixture was added DCM (500 mL) followed by (BOC)20 (36.4 g, 166.97
mmol). The
biphasic reaction mixture was stirred vigorously overnight. The phases were
then separated
and the aqueous layer was extracted with DCM (500 mL x 5). The combined
organic phases
dried (Na2SO4), filtered and the solvent was evaporated under reduced
pressure. The material
was purified by chromatography using a 120 g silica cartridge eluting with
heptanes ¨ Et0Ac
(gradient 0% to 30% Et0Ac) to give. tert-butyl 2-(tert-
butoxycarbonyl(trideuteriomethyl)amino)acetate (6 g, 24.19 mmol, 28% yield) as
a colorless
oil. 1H NMR (CDC13) 3: 3.84-3.74 (m, 2H), 1.45-1.41 (m, 18H).
Step 4: Synthesis of tert-butyl 2-(N-trideuterio methylamino)acetate TFA salt
TFA
HN-Thr
CD3 0
[00381] A round bottom flask equipped with a stir bar was charged with tert-
butyl 2-
(tert-butoxy carb onyl(tri deuteri omethyDamino)acetate (500 mg, 2.02 mmol)
and
dichloromethane (2 mL). The mixture was cooled to 0 C at which time 1 mL of
trifluoroacetic acid (TFA) was added. The mixture was then stirred at 0 C for
3 hours. The
123

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
solvent was then evaporated under reduced pressure to give tert-butyl 2-(N-
trideuterio
methylamino)acetate TFA salt (320 mg, 2.02 mmol, 100% yield) as a pale brown
oil, which
was used directly in the next step. ES LC-MS m/z = 149 (M+H+).
Step 5: Synthesis of tert-butyl 2-(N-trideuteriomethylcyanamido)acetate
CD3 0
[00382] A round bottom flask equipped with a stir bar was charged with tert-
butyl 2-
(N-trideuterio methylamino)acetate TFA salt (320mg, 2.02mm01), potassium
carbonate (837
mg, 6.06 mmol) and acetonitrile (10 mL). The mixture was stirred at room
temperature for
0.5 hours, at which time a solution of cyanogen bromide (235 mg, 2.22 mmol) in
acetonitrile
(2 ml) was added. The reaction was allowed to stir overnight at room
temperature. The
solvent was then decanted leaving behind an insoluble residue. The solvent was
evaporated
under reduced pressure and then purified by flash chromatography eluting with
petroleum
ether - ethyl acetate (gradient 0% to 40% ethyl acetate) to give tert-butyl 2-
(N-
trideuteriomethylcyanamido)acetate (260 mg, 1.50 mmol, 74% yield) as a white
solid. ES
LC-MS m/z = 174 (M+H+) & 196 (M+Na+).
Step 6: Synthesis of tert-butyl 2-(2-hydroxy-1-
trideuteriomethylguanidino)acetate
N,OH
H2NILNThr0..,.-
CD3 0
[00383] A round bottom flask equipped with a stir bar was charged with ter/-
butyl 2-
(N-trideuteriomethylcyanamido)acetate (260 mg, 1.50 mmol) and tetrahydrofuran
(5 mL). To
this mixture was added hydroxylamine (50% aqueous, 495 mg, 7.50 mmol). After 1
hour, 10
mL of water was added and the mixture was extracted with 5 mL of
dichloromethane 3 times.
The organic layer was dried with MgSO4, filtered and concentrated under
reduced pressure to
give tert-butyl 2-(2-hydroxy-1-trideuteriomethylguanidino)acetate (298 mg,
1.45 mmol, 97%
yield) as a white solid which was used immediately in the next step. ES LC-MS
m/z = 207
(M+H+).
Step 7: Synthesis of tert-butyl 2-(3-(tert-butoxycarbony1)-2-hydroxy-l-
trideuteriomethylguanidino)acetate
124

CA 02971729 2017-06-20
WO 2016/106284 PCT/1JS2015/067283
N_OH
Boc,N)1,N,,,i0-
H
CD3 0
[00384] A round bottom flask equipped with a stir bar was charged with tert-
butyl 2-
(2-hydroxy-1-trideuteriomethylguanidino)acetate (298 mg, 1.45 mmol) and
tetrahydrofuran
(5 mL). To this mixture was added di-tert-butyl dicarbonate (316 mg, 1.72
mmol). The
reaction was stirred at room temperature overnight. The solvent was then
evaporated under
reduced pressure and the material was purified by flash chromatography eluting
with
dichloromethane - ethyl acetate (gradient 0% to 30% ethyl acetate) to give
tert-butyl 2-(3-
(tert-butoxycarbony1)-2-hydroxy-1-trideuteriomethylguanidino)acetate (90 mg,
0.29 mmol,
20% yield) as a white solid. ES LC-MS m/z = 307 (M+H+). 11-1 NMR (DIMETHYL
SULFOXIDE-d) 5: 5.71 (s, 2H), 3.81 (s, 2H), 1.43 (s, 9H), 1.41 (s, 9H).
[00385] Other creatine prodrugs and derivatives thereof can be synthesized
using the
procedure described above by the selection of the appropriate starting
material.
Example 26: Synthesis of ethyl 2-[methyl-(5-oxo-4H-1,2,4-oxadiazol-3-
yl)amino]acetate
i) NH2OH.HCI, Et3N
N THF, RT, 18 h O-N
, C)
N õ
I 8
ii) CD!, RI, 1 h NII
H 0
[00386] A round bottom flask equipped with a stir bar and nitrogen inlet
was charged
with ethyl 2-[cyano(methyl)amino]acetate (852 mg, 6.0 mmol) and
tetrahydrofuran (30 mL).
To this mixture was added hydroxylamine hydrochloride (2.1 g, 30.0 mmol) and
triethylamine (1.3 mL, 9.0 mmol). After 18 h, carbonyldiimide (5.8 g, 36.0
mmol) was added
and the reaction was allowed 1 h. The solvent was decanted leaving behind an
insoluble
residue. The solvent was evaporated under reduced pressure and the material
was purified by
reverse phase chromatography eluting with water-acetonitrile modified with
0.1%
trifluoroacetic acid. This gave ethyl 2-[methyl-(5-oxo-4H-1,2,4-oxadiazol-3-
yl)amino]acetate
as a white solid: 80 mg, 0.40 mmol, 7% yield. ES LC-MS m/z = 202 (M+H+). 1-1-1
NMR
(CHLOROFORM-d) 6: 425 (q, ./ = 7.2 Hz, 2H), 3.96 (s, 2H), 3.04 (s, 3H), 1.31
(t, .1 = 7.1
Hz, 3H). Melting point 120-123 C.
[00387] Eethyl 2-[methyl-(5-oxo-4H-1,2,4-oxadiazol-3-yl)amino]acetate can
also be
synthesized using the procedure described in Kitamure et al, Chem. Pharm.
Bull,, 2001, 49(3)
268-277.
125

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
Example 27: Synthesis of 2-[methyl-(5-oxo-4H-1,2,4-oxadiazol-3-yl)amino]acetic
acid
O¨N LION, THF, H20
NN
N
JLNO H I RT, 1 h JLH
OH
' 0 ' 0
[00388] A round bottom flask equipped with a stir bar and nitrogen inlet
was charged
with ethyl 2-[methyl-(5-oxo-4H-1,2,4-oxadiazol-3-yl)amino]acetate (20 mg, 0.1
mmol),
tetrahydrofuran (5 mL) and water (5 mL). To this mixture was added lithium
hydroxide mono
hydrate (4 mg, 0.1 mmol). After 1 hour the solvent was evaporated under
reduced pressure
and the material was purified by reverse phase chromatography eluting with
water¨
acetoni trile modified with 0.1% trifluoroaceti c acid. This gave 2-[m ethyl -
(5 -ox o-4H-1,2,4-
oxadiazol-3-yl)amino]acetic acid as a white solid: 15 mg, 0.09 mmol, 90%
yield. ES LC-MS
m/z = 174 (M+H+). 11-1 NMR (METHANOL-d4) 6: 3.97 (s, 2H), 2.96 (s, 3H).
Melting point
150-155 C.
[00389] Other creatine prodrugs and derivatives thereof can be synthesized
using the
procedure described above by the selection of the appropriate starting
material.
Example 28: Synthesis of alkyl 2-[(5-oxo-2H-1,2,4-oxadiazol-3-y1)-
(trideuteriomethypamino]acetate
Step 1
Step 2
ethyl N-dhioxomethylene)
TFA. 0 S NaH, Mel, THF 0 S
0 carbamate
1-111.1 0-&1\Na
(.-L
CD3 0 CH2Cl2, 0 C to RT H I RT, 30 min
CD3 0 O
18 h D3 0
Step 3 Step 4
Step 5
Li0H.H20, THF R-OH, EDC,
Hydroxylamine.HCI 0¨NH CD¨NH
pyridine H20 DMAP, Et3N 0\
N
50 C, 1 h OD3 0 RT, 1 h OD3 0 CH2Cl2, RT, 2 h OD3 0
R = heptyl, ethyl, iPr
Step 1: Synethsis of methyl 2-
[ethoxy carb onyl carb am othi oyl (tri deuteri omethyl)amino]acetate
0 S
0
H
0
D D
[00390] A round bottom flask equipped with a stir bar and nitrogen inlet
was charged
with ethyl N-(thioxomethylene)carbamate (4.1 mL, 35.0 mmol) and
dichloromethane (300
126

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
mL). The mixture was cooled to 0 C and methyl 2-
(trideuteriomethylamino)acetate
trifluoroacetic acid salt (7.70 g, 35.0 mmol) was added followed by
triethylamine (4.9 mL,
35.0 mmol). The mixture was stirred overnight with warming to room
temperature. The
mixture was washed with 1N HC1 (100 mL), dried (Na2SO4) and the solvent was
evaporated
under reduced pressure. The material was purified by chromatography using a
120 g silica
cartridge eluting with heptane¨ethyl acetate, gradient 0 to 30% ethyl acetate.
This gave
methyl 2-[ethoxyc arb onyl carb am othi oyl(tri deuteri omethyl)amino] acetate
: 8.0 g, 33.8 mmol,
96% yield. ES LC-MS m/z = 238 (M+H-). 1H NMR (CHLOROFORM-d) 6: 7.38 (br s,
1H),
4.41-4.59 (m, 2H), 4.20 (q, J= 7.1 Hz, 2H), 3.80 (s, 3H), 1.31 (t, J= 7.1 Hz,
3H).
Step 2: Synethsis of methyl 2-[[(Z)-N-ethoxycarbonyl-C-methylsulfanyl-
carbonimidoy1]-
(tri deuteri omethypamino] acetate
0 S
D
)(DD
0
[00391] A round bottom flask equipped with a stir and nitrogen inlet was
charged with
[ethoxy carb onyl carb am othi oyl (tri deuteri omethypamino]acetate (7.82 g,
33.0 mmol),
methyliodide (4.1 mL, 66.0 mmol) and tetrahydrofuran (200 mL). To this mixture
at room
temperature was added sodium hydride (60% in oil; 1.32 g, 33.0 mmol). After 1
hour, the
material was poured into saturated ammonium chloride solution (100 mL), the
aqueous phase
was extracted with ethyl acetate (3 X 100 mL), dried (Na2SO4) and the solvent
was
evaporated under reduced pressure. The material was purified by chromatography
using a
120 g silica cartridge eluting with heptane¨ethyl acetate, gradient 0 to 40%
ethyl acetate. This
gave methyl 2-[[(Z)-N-ethoxycarbonyl-C-methylsulfanyl-
carbonimidoy1]-
(tri deuteriomethyl)amino]acetate: 8.0 g, 32.0 mmol, 97% yield. ES LC-MS m/z =
252
(M+H+). 1H NMR (CHLOROFORM-d) 6: 4.30 (s, 2H), 4.16 (q, J= 7.1 Hz, 2H), 3.77
(s, 3H),
2.42 (s, 3H), 1.30 (t, J= 7.1 Hz, 3H).
Step 3: Synethsis of methyl 2-[(5-oxo-2H-1,2,4-oxadiazol-3-y1)-
(trideuteriomethyl)amino]acetate
C)I-NH
0 õA,
N
.....---., 0
D D
127

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
[00392] A round
bottom flask equipped with a stir bar, Vigreux column and nitrogen
inlet was charged with methyl 2-[[(Z)-N-ethoxycarbonyl-C-methylsulfanyl-
carbonimidoy1]-
(trideuteriomethyDamino]acetate (7.53 g, 30.0 mmol) and pyridine (50 mL). To
this mixture
was added hydroxylamine hydrochloride (2.09 g, 30.0 mmol) and the mixture was
heated at
60 C for 1 hour. The solvent was evaporated under reduced pressure. Ethyl
acetate (100 mL)
and water (100 mL) was added. The phases were separated and the aqueous phase
was
extracted with ethyl acetate (3 X 100 mL). The combined organic phases were
dried
(Na2SO4) and the solvent was evaporated under reduced pressure. The material
was purified
by chromatography using a 120 g silica cartridge eluting with heptane¨ethyl
acetate, gradient
0 to 100% ethyl acetate. This gave methyl 2-[(5-oxo-2H-1,2,4-oxadiazol-3-y1)-
(trideuteriomethypamino]acetate as a white solid : 3.0 g, 15.8 mmol, 53%
yield. ES LC-MS
m/z = 191 (M+TF). 1H NMR (CHLOROFORM-d) 6: 3.98 (s, 2H), 3.79 (s, 3H). Melting
point
120-125 C.
Step 4: Synethsis of 2-[(5-oxo-2H-1,2,4-oxadiazol-3-y1)-
(trideuteriomethypamino]acetic acid
(:)¨NH
N N,ThrOH
0
D D
[00393] A round
bottom flask equipped with a stir bar and nitrogen inlet was charged
with methyl 2-[(5-oxo-2H-1,2,4-oxadiazol-3-y1)-
(trideuteriomethyl)amino]acetate (95 mg,
0.5 mmol), tetrahydrofuran (5 mL) and water (5 mL). To this mixture was added
lithium
hydroxide mono hydrate (21 mg, 0.5 mmol). After 1 hour the solvent was
evaporated under
reduced pressure and the material was purified by reverse phase chromatography
eluting with
water¨acetonitrile modified with 0.1% trifluoroacetic acid. This gave 2-[(5-
oxo-2H-1,2,4-
oxadiazol-3-y1)-(trideuteriomethyl)amino]acetic acid as a white solid : 50 mg,
0.28 mmol,
57% yield. ES LC-MS m/z = 177 (M+H+). 11-1 NMR (METHANOL-d4) 6. 3.98 (s, 2H).
Melting point 150-155 C.
Step 5A: Synethsis of heptyl 2- [(5 -
oxo-2H-1,2,4-oxadiazol-3 -y1)-
(trideuteriomethypamino]acetate
ID-NH
0
N N
0
128

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
[00394] A
scintillation vial equipped with a stir bar was charged with 2-[(5-oxo-2H-
1,2,4-oxadiazol-3-y1)-(trideuteriomethyl)amino]acetic acid (88 mg, 0.50 mmol),
1-heptanol
(58 mg, 0.50 mmol), dimethylamino pyridine (92 mg, 0.75 mmol) and
dichloromethane (10
mL). To this mixture was added N-(3-dimethylaminopropy1)-N'-ethylcarbodiimide
hydrochloride (144 mg, 0.75 mmol). After 1 hour, triethylamine (0.07 mL, 0.50
mmol) was
added. The reaction was allowed another 1 hour. The solvent was evaporated
under reduced
pressure and the material was purified by reverse phase chromatography eluting
with water¨
acetonitrile modified with 0.1% trifluoroacetic acid. This gave heptyl 2-[(5-
oxo-2H-1,2,4-
oxadiazol-3-y1)-(trideuteriomethypamino]acetate as a white solid : 70 mg, 0.25
mmol, 50%
yield. ES LC-MS m/z = 275 (M+H+). 1H NMR (CHLOROFORM-d) 6: 11.06 (br s, 1H),
4.17
(t, ,1= 6.8 Hz, 2H), 3.95 (s, 2H), 1.56-1.82 (m, 4H), 1.19-1.44 (m, 7H), 0.85-
0.94 (m, 2H).
Melting point 110-115 C.
Step 5B: Synethsi s of ethyl 2-[(5-
oxo-2H-1,2,4-oxadiazol-3-y1)-
(trideuteri omethyl)amino] acetate
"-NH
N
.D
D D
[00395] A
scintillation vial equipped with a stir bar was charged with 2-[(5-oxo-2H-
1,2,4-oxadiazol-3-y1)-(trideuteriomethyl)amino]acetic acid (176 mg, 1.00
mmol), ethanol (46
mg, 1.00 mmol), dimethylamino pyridine (183 mg, 1.50 mmol) and dichloromethane
(20
mL). To this mixture was added N-(3-dimethylaminopropy1)-N'-ethylcarbodiimide
hydrochloride (288 mg, 1.50 mmol). After 1 hour, triethylamine (0.14 mL, 1.00
mmol) was
added. The reaction was allowed another 1 hour. The solvent was evaporated
under reduced
pressure and the material was purified by reverse phase chromatography eluting
with water¨
acetonitrile modified with 0.1% trifluoroacetic acid. This gave ethyl 2-[(5-
oxo-2H-1,2,4-
oxadiazol-3-y1)-(trideuteriomethypamino]acetate as a white solid : 133 mg,
0.65 mmol, 65%
yield. ES LC-MS m/z = 205 (M+H+). 1H NMR (CHLOROFORM-d) 6: 11.13 (br s, 1H),
4.24
(q, J= 7.2 Hz, 2H), 3.96 (s, 2H), 1.30 (t, J=7.1 Hz, 3H). Melting point 12-130
C.
Step 5C: Synethsi s of isopropyl 2- [(5 -
oxo-2H-1,2,4-oxadiazol-3 -y1)-
(tri deuteri om ethyl )am i n o] acetate
129

CA 02971729 2017-06-20
WO 2016/106284 PCT/US2015/067283
-NH
N
D
,J<DD 011
[00396] A scintillation vial equipped with a stir bar was charged with 2-
[(5-oxo-2H-
1,2,4-oxadiazol-3-y1)-(trideuteriomethyl)amino]acetic acid (176 mg, 1.00
mmol), isopropanol
(60 mg, 1.00 mmol), dimethylamino pyridine (183 mg, 1.50 mmol) and
dichloromethane (20
mL). To this mixture was added N-(3-dimethylaminopropy1)-N'-ethylcarbodiimide
hydrochloride (288 mg, 1.50 mmol). After 1 hour, triethylamine (0.14 mL, 1.00
mmol) was
added. The reaction was allowed another 1 hour. The solvent was evaporated
under reduced
pressure and the material was purified by reverse phase chromatography eluting
with water¨
acetonitrile modified with 0.1% trifluoroacetic acid. This gave isopropyl 2-
[(5-oxo-2H-1,2,4-
oxadiazol-3-y1)-(trideuteriomethypamino]acetate as a white solid : 139 mg,
0.64 mmol, 64%
yield. ES LC-MS m/z = 219 (M+H+). 1H NMR (CHLOROFORM-d) 6: 11.09 (br s, 1H),
5.10
(sept, J= 6.3 Hz, 1H), 3.91 (s, 2H), 1.28 (d, J= 6.2 Hz, 6H). Melting point
143-145 C.
Example 29: Synthesis of 3- [2-hy droxy ethyl (tri deuteri om ethyl)am i no] -
2H-1,2,4-oxadi azol -
5-one
0- BHITHF, THF
NH - 0 C to RT NH
N
N N
CD3 0 CD3
[00397] A round bottom flask equipped with a stir bar and nitrogen inlet
was charged
with 2-[(5-oxo-2H-1,2,4-oxadiazol-3-y1)-(trideuteriomethypamino]acetic acid
(528 mg, 3.0
mmol) and tetrahydrofuran (Ti-IF) (60 mL). To this mixture was added BH3.T1-IF
(1.0 M in
THF) (6.0 mL, 6.0 mmol). After 2 h, the reaction was quenched with methanol (5
mL) and
the solvent was evaporated under reduced pressure. The material was purified
by reverse
phase chromatography eluting with water¨acetonitrile modified with 0.1%
trifluoroacetic
acid. This gave 3-[2-hydroxyethyl(trideuteriomethyl)amino]-2H-1,2,4-oxadiazol-
5-one as a
white solid : 372 mg, 2.30 mmol, 77% yield. ES LC-MS m/z = 163 (M+H+). 11-1
NMR
(METHANOL-d4) 6: 3.70 (t, J= 5.4 Hz, 3H), 3.33 (s, 1H). Melting point 98-104
C.
Example 30: Synthesis of 3-imino-4-(methyl-d3)-1,2,4-oxadiazinan-6-one and 3-
amino-4-
(methyl-d3)-4,5 -di hy dro-6H-1,2,4-oxadi azin-6-one
130

CA 02971729 2017-06-20
WO 2016/106284
PCT/1JS2015/067283
Step 1
Step 2 r0 0,
H BrCN 0 NH2OH. HCI
N
K2CO3, Me0,
D N 11
0 AcONa, Me0H, NH + NH2
D HCI 25 C, 5 h
25 C, 4 h DD DAD
Step 1: Synthesis of methyl 2-[cyano(trideuteriomethyl)amino]acetate
111 0
Erl
[00398] A flask
was fitted with methyl 2-(trideuteriomethylamino) acetate (3.00 g,
21.04 mmol, HC1 salt), K2CO3 (5.82 g, 42.08 mmol) and Me0H (20.00 mL). Then
carbononitridic bromide (2.23 g, 21.04 mmol) was added. After that the
reaction mixture was
stirred for about 5 hour at 25 C. The reaction mixture was evaporated to
afford the residue.
H20 (20 mL) was added and EtOAc (35 mL X 5) was used to extract the product.
The
organic layer was washed by brine (20 mL), dried over anhydrous Na2SO4 and
evaporated to
afford methyl 2-[cyano(trideuteriomethyl)amino]acetate (2.00 g, 72.48% yield)
as yellow red
oil. 1H NMR (Me0D, 400 MHz) 6: 3.91 (s, 2H), 3.79 (s, 3H).
Step 2: Synthesis of 3-imino-4-(methyl-d3)-1,2,4-oxadiazinan-6-one and 3-amino-
4-(methyl-
0-4,5 -di hy dro-6H-1,2,4-oxadi azin-6-one
NNH
NH N
+ N NH2
DD DkD
[00399] A flask
was fitted with methyl 2-[cyano(trideuteriomethyl)amino]acetate (1.00
g, 7.62 mmol), hydroxylamine (2.12 g, 30.50 mmol, HC1 salt) and AcONa (2.81 g,
34.31
mmol) in Me0H (20.00 mL). The reaction mixture was stirred at 25 C for about
4 h. The
reaction solution was evaporated to remove most of Me0H. Then H20 (about 3 mL)
was
added to make all the solid dissolve. Half of the solution was directly
purified by special
prep-HPLC (neutral) to afford 4 peaks with desired MS. Peak C, 39.7 mg, light
pink. ES LC-
MS m/z = 130 (M+H+). 1H NMR (DMSO-d6) 6: 3.67 (s, 2H). Peak D, 70.7 mg, white
solid.
ES LC-MS m/z = 130 (M+H-). 114 NMR (DMSO-d6) 6: 3.64 (s, 2H). The remaining
peaks
may correspond to 3 -hy droxy -2-
imino-1 -(methyl-dl)imi dazol i din-4 -one, (Z)-2-
131

(hydroxyimino)-1 -(m ethyl-d3)imi daz oli din-4-one, or (E)-2-(hydroxyimino)-1
-(m ethyl -d3)imi daz oli din-
4-one.
Example 31: Synthesis of 4-amino-2,5-dimethy1-5,6-dihydro-1,3,5-oxadiazepin-
7(21/)-one
NH2
0
0 k
H
OH TFA, Dioxane, ref lux N
N N H2
0 propa n-2-one
[00400] The title compound, 4-amino-2,5-dimethy1-5,6-dihydro-1,3,5-
oxadiazepin-7(21/)-one,
can be synthesized from the readily available starting materials creatine
(available from Sigma Aldrich)
and propan-2-one (acetone, also available from Sigma Aldrich). To a stirred
solution of creatine (6.55
g; 50 mmol) and acetone (3.70 mL, 50 mmol) in anhydrous dioxane (100 mL) is
added trifloroacetic acid
(TFA; 2.0 mL; 26 mmol) over 10 minutes. The solution is stirred at room
temperature for 10 min,
followed by reflux for 1 h, then concentrated in vacuo to yield the crude
product. The crude product is
dissolved in a minimum amount of anhydrous dioxane followed by introduction of
a stream of anhydrous
HC1 gas. The resulting precipitate is filtered and washed with dry diethyl
ether to yield the HC1 salt of
the title compound.
[00401] By using other aldehydes or ketone instead of propan-2-one, other
4-amino-5,6-dihydro-
1,3,5-oxadiazepin-7(21/)-one compounds can be prepared.
[00402] Other creatine prodrugs can be synthesized using the procedure
described above by the
selection of the appropriate starting material.
[00403] The invention can be embodied in other specific forms without
departing from the spirit
or essential characteristics thereof. The foregoing embodiments are therefore
to be considered in all
respects illustrative rather than limiting on the invention described herein.
Scope of the invention is thus
indicated by the appended claims rather than by the foregoing description, and
all changes that come
within the meaning and range of equivalency of the claims are intended to be
embraced therein.
132
Date Recue/Date Received 2022-04-14

Dessin représentatif
Une figure unique qui représente un dessin illustrant l'invention.
États administratifs

2024-08-01 : Dans le cadre de la transition vers les Brevets de nouvelle génération (BNG), la base de données sur les brevets canadiens (BDBC) contient désormais un Historique d'événement plus détaillé, qui reproduit le Journal des événements de notre nouvelle solution interne.

Veuillez noter que les événements débutant par « Inactive : » se réfèrent à des événements qui ne sont plus utilisés dans notre nouvelle solution interne.

Pour une meilleure compréhension de l'état de la demande ou brevet qui figure sur cette page, la rubrique Mise en garde , et les descriptions de Brevet , Historique d'événement , Taxes périodiques et Historique des paiements devraient être consultées.

Historique d'événement

Description Date
Inactive : Octroit téléchargé 2024-03-06
Inactive : Octroit téléchargé 2024-03-04
Inactive : Octroit téléchargé 2024-03-04
Inactive : Octroit téléchargé 2024-02-26
Accordé par délivrance 2024-02-20
Lettre envoyée 2024-02-20
Inactive : Page couverture publiée 2024-02-19
Paiement d'une taxe pour le maintien en état jugé conforme 2024-02-13
Inactive : Taxe finale reçue 2023-12-29
Préoctroi 2023-12-29
Lettre envoyée 2023-12-22
month 2023-09-18
Lettre envoyée 2023-09-18
Un avis d'acceptation est envoyé 2023-09-18
Inactive : Approuvée aux fins d'acceptation (AFA) 2023-07-24
Inactive : Q2 réussi 2023-07-24
Modification reçue - réponse à une demande de l'examinateur 2023-03-10
Modification reçue - modification volontaire 2023-03-10
Rapport d'examen 2022-11-17
Inactive : Rapport - Aucun CQ 2022-10-31
Modification reçue - modification volontaire 2022-04-14
Modification reçue - réponse à une demande de l'examinateur 2022-04-14
Rapport d'examen 2021-12-17
Inactive : Rapport - Aucun CQ 2021-12-16
Représentant commun nommé 2020-11-07
Lettre envoyée 2020-10-21
Exigences pour une requête d'examen - jugée conforme 2020-10-13
Toutes les exigences pour l'examen - jugée conforme 2020-10-13
Requête d'examen reçue 2020-10-13
Représentant commun nommé 2019-10-30
Représentant commun nommé 2019-10-30
Requête visant le maintien en état reçue 2018-12-07
Requête visant le maintien en état reçue 2017-12-04
Inactive : Page couverture publiée 2017-11-15
Inactive : Notice - Entrée phase nat. - Pas de RE 2017-07-06
Inactive : CIB en 1re position 2017-06-30
Inactive : CIB attribuée 2017-06-30
Inactive : CIB attribuée 2017-06-30
Inactive : CIB attribuée 2017-06-30
Inactive : CIB attribuée 2017-06-30
Inactive : CIB attribuée 2017-06-30
Inactive : CIB attribuée 2017-06-30
Inactive : CIB attribuée 2017-06-30
Inactive : CIB attribuée 2017-06-30
Demande reçue - PCT 2017-06-30
Exigences pour l'entrée dans la phase nationale - jugée conforme 2017-06-20
Demande publiée (accessible au public) 2016-06-30

Historique d'abandonnement

Il n'y a pas d'historique d'abandonnement

Taxes périodiques

Le dernier paiement a été reçu le 2024-02-13

Avis : Si le paiement en totalité n'a pas été reçu au plus tard à la date indiquée, une taxe supplémentaire peut être imposée, soit une des taxes suivantes :

  • taxe de rétablissement ;
  • taxe pour paiement en souffrance ; ou
  • taxe additionnelle pour le renversement d'une péremption réputée.

Les taxes sur les brevets sont ajustées au 1er janvier de chaque année. Les montants ci-dessus sont les montants actuels s'ils sont reçus au plus tard le 31 décembre de l'année en cours.
Veuillez vous référer à la page web des taxes sur les brevets de l'OPIC pour voir tous les montants actuels des taxes.

Historique des taxes

Type de taxes Anniversaire Échéance Date payée
Taxe nationale de base - générale 2017-06-20
TM (demande, 2e anniv.) - générale 02 2017-12-22 2017-12-04
TM (demande, 3e anniv.) - générale 03 2018-12-24 2018-12-07
TM (demande, 4e anniv.) - générale 04 2019-12-23 2019-12-13
Requête d'examen - générale 2020-12-22 2020-10-13
TM (demande, 5e anniv.) - générale 05 2020-12-22 2020-12-18
TM (demande, 6e anniv.) - générale 06 2021-12-22 2021-12-17
TM (demande, 7e anniv.) - générale 07 2022-12-22 2022-12-16
Pages excédentaires (taxe finale) 2023-12-29 2023-12-29
Taxe finale - générale 2023-12-29
TM (demande, 8e anniv.) - générale 08 2023-12-22 2024-02-13
Surtaxe (para. 27.1(2) de la Loi) 2024-02-13 2024-02-13
Titulaires au dossier

Les titulaires actuels et antérieures au dossier sont affichés en ordre alphabétique.

Titulaires actuels au dossier
FARMINGTON PHARMA DEVELOPMENT
Titulaires antérieures au dossier
WILLIAM F. BRUBAKER
Les propriétaires antérieurs qui ne figurent pas dans la liste des « Propriétaires au dossier » apparaîtront dans d'autres documents au dossier.
Documents

Pour visionner les fichiers sélectionnés, entrer le code reCAPTCHA :



Pour visualiser une image, cliquer sur un lien dans la colonne description du document (Temporairement non-disponible). Pour télécharger l'image (les images), cliquer l'une ou plusieurs cases à cocher dans la première colonne et ensuite cliquer sur le bouton "Télécharger sélection en format PDF (archive Zip)" ou le bouton "Télécharger sélection (en un fichier PDF fusionné)".

Liste des documents de brevet publiés et non publiés sur la BDBC .

Si vous avez des difficultés à accéder au contenu, veuillez communiquer avec le Centre de services à la clientèle au 1-866-997-1936, ou envoyer un courriel au Centre de service à la clientèle de l'OPIC.


Description du
Document 
Date
(yyyy-mm-dd) 
Nombre de pages   Taille de l'image (Ko) 
Page couverture 2024-01-18 1 44
Dessin représentatif 2024-01-18 1 5
Revendications 2023-03-09 14 496
Description 2017-06-19 132 6 862
Revendications 2017-06-19 20 530
Abrégé 2017-06-19 1 63
Page couverture 2017-08-30 1 37
Description 2022-04-13 132 7 002
Revendications 2022-04-13 14 350
Abrégé 2023-03-09 1 33
Taxe finale 2023-12-28 4 116
Paiement de taxe périodique 2024-02-12 1 29
Certificat électronique d'octroi 2024-02-19 1 2 527
Avis d'entree dans la phase nationale 2017-07-05 1 192
Rappel de taxe de maintien due 2017-08-22 1 113
Courtoisie - Réception de la requête d'examen 2020-10-20 1 437
Avis du commissaire - Demande jugée acceptable 2023-09-17 1 578
Courtoisie - Réception du paiement de la taxe pour le maintien en état et de la surtaxe 2024-02-12 1 422
Avis du commissaire - non-paiement de la taxe de maintien en état pour une demande de brevet 2024-02-01 1 552
Paiement de taxe périodique 2018-12-06 1 40
Rapport de recherche internationale 2017-06-19 3 149
Demande d'entrée en phase nationale 2017-06-19 3 92
Déclaration 2017-06-19 1 13
Paiement de taxe périodique 2017-12-03 1 40
Requête d'examen 2020-10-12 4 109
Demande de l'examinateur 2021-12-16 6 293
Modification / réponse à un rapport 2022-04-13 73 3 245
Demande de l'examinateur 2022-11-16 4 191
Modification / réponse à un rapport 2023-03-09 35 934